1
|
Mi Y, Yu H, Wang P, Miao Y, Teng X, Jin S, Xiao L, Xue H, Tian D, Guo Q, Wu Y. Tyrosine hydroxylase-positive neurons in the rostral ventrolateral medulla mediate sympathetic activation in sepsis. Life Sci 2024; 358:123118. [PMID: 39384147 DOI: 10.1016/j.lfs.2024.123118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 09/26/2024] [Accepted: 10/05/2024] [Indexed: 10/11/2024]
Abstract
AIM Sepsis results in high mortality and is associated with organ dysfunction caused by infection. The present study aimed to elucidate whether early-stage sympathetic activation is associated with the prognosis of sepsis and its possible mechanisms. METHODS Patients with sepsis and healthy controls were included. Sepsis in rats was induced by lipopolysaccharide. Dexmedetomidine, a α2-adrenergic receptor agonist, was used in patients and rats with sepsis to evaluate the role of the sympathetic nervous system in sepsis. Holter monitoring was used to detect heart rate variability, while plasma samples were obtained to measure levels of norepinephrine and inflammatory markers. Mean arterial pressure, heart rate, and renal sympathetic nerve activity were recorded. Immunofluorescence was used to detect the activation of neurons in the rostral ventrolateral medulla (RVLM). RESULTS In patients with sepsis, plasma levels of norepinephrine and interleukin-1β were higher compared with those in controls and positively correlated with acute physiology and chronic health evaluation (APACHE) II. SDNN and SDANN were significantly reduced as well as negatively correlated with APACHE II. Meanwhile, rats with sepsis showed increased of sympathetic outflow and plasma levels of norepinephrine, with increased c-fos levels in the RVLM. Treatment with dexmedetomidine could improve prognosis. Lesion of tyrosine hydroxylase-positive neurons in the RVLM attenuated sympathetic activation and target organs damage in septic rats as well as improved survival. CONCLUSION The results suggest that tyrosine hydroxylase-positive neurons in the RVLM might contribute to the prognosis of sepsis via activation of the sympathetic nervous system, while dexmedetomidine could ameliorate sepsis via inhibiting sympathetic activation.
Collapse
Affiliation(s)
- Yuan Mi
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China; Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Hao Yu
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Ping Wang
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Yuxin Miao
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Xu Teng
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Sheng Jin
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Lin Xiao
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Hongmei Xue
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Danyang Tian
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Qi Guo
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China; Experimental Center for Teaching, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, 050017 Hebei, China.
| | - Yuming Wu
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China; Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang 050017, China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, 050017 Hebei, China.
| |
Collapse
|
2
|
Hornung E, Achanta S, Moss A, Schwaber JS, Vadigepalli R. Multi-organ gene expression analysis and network modeling reveal regulatory control cascades during the development of hypertension in female spontaneously hypertensive rat. PLoS One 2024; 19:e0313252. [PMID: 39514592 PMCID: PMC11548744 DOI: 10.1371/journal.pone.0313252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Hypertension is a multifactorial disease with stage-specific gene expression changes occurring in multiple organs over time. The temporal sequence and the extent of gene regulatory network changes occurring across organs during the development of hypertension remain unresolved. In this study, female spontaneously hypertensive (SHR) and normotensive Wistar Kyoto (WKY) rats were used to analyze expression patterns of 96 genes spanning inflammatory, metabolic, sympathetic, fibrotic, and renin-angiotensin (RAS) pathways in five organs, at five time points from the onset to established hypertension. We analyzed this multi-dimensional dataset containing ~15,000 data points and developed a data-driven dynamic network model that accounts for gene regulatory influences within and across visceral organs and multiple brainstem autonomic control regions. We integrated the data from female SHR and WKY with published multiorgan gene expression data from male SHR and WKY. In female SHR, catecholaminergic processes in the adrenal gland showed the earliest gene expression changes prior to inflammation-related gene expression changes in the kidney and liver. Hypertension pathogenesis in male SHR instead manifested early as catecholaminergic gene expression changes in brainstem and kidney, followed by an upregulation of inflammation-related genes in liver. RAS-related gene expression from the kidney-liver-lung axis was downregulated and intra-adrenal RAS was upregulated in female SHR, whereas the opposite pattern of gene regulation was observed in male SHR. We identified disease-specific and sex-specific differences in regulatory interactions within and across organs. The inferred multi-organ network model suggests a diminished influence of central autonomic neural circuits over multi-organ gene expression changes in female SHR. Our results point to the gene regulatory influence of the adrenal gland on spleen in female SHR, as compared to brainstem influence on kidney in male SHR. Our integrated molecular profiling and network modeling identified a stage-specific, sex-dependent, multi-organ cascade of gene regulation during the development of hypertension.
Collapse
Affiliation(s)
- Eden Hornung
- Department of Pathology and Genomic Medicine, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Sirisha Achanta
- Department of Pathology and Genomic Medicine, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Alison Moss
- Department of Pathology and Genomic Medicine, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - James S. Schwaber
- Department of Pathology and Genomic Medicine, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Rajanikanth Vadigepalli
- Department of Pathology and Genomic Medicine, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
3
|
Lo J, Melhorn SJ, Kee S, Olerich KLW, Huang A, Yeum D, Beiser A, Seshadri S, De Carli C, Schur EA. Hypothalamic Gliosis is Associated With Multiple Cardiovascular Disease Risk Factors. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.19.24313914. [PMID: 39371136 PMCID: PMC11451704 DOI: 10.1101/2024.09.19.24313914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Background Hypothalamic gliosis is mechanistically linked to obesity and insulin resistance in rodent models. We tested cross-sectional associations between radiologic measures of hypothalamic gliosis in humans and clinically relevant cardiovascular disease risk factors, as well as prevalent coronary heart disease. Methods Using brain MRI images from Framingham Heart Study participants (N=867; mean age, 55 years; 55% females), T2 signal intensities were extracted bilaterally from the region of interest in the mediobasal hypothalamus (MBH) and reference regions in the amygdala (AMY) and putamen (PUT). T2 signal ratios were created in which greater relative T2 signal intensity suggests gliosis. The primary measure compared MBH to AMY (MBH/AMY); a positive control ratio (MBH/PUT) also assessed MBH whereas a negative control (PUT/AMY) did not. Outcomes were BMI, HDL-C, LDL-C, fasting triglycerides, and the presence of hypertension (n=449), diabetes mellitus (n=66), metabolic syndrome (n=254), or coronary heart disease (n=25). Dietary risk factors for gliosis were assessed in a prospective analysis. Statistical testing was performed using linear or logistic regression. Results Greater MBH/AMY T2 signal ratios were associated with higher BMI (β = 21.5 [95% CI, 15.4-27.6]; P<0.001), higher fasting triglycerides (β = 1.1 [95% CI, 0.6-1.7]; P<0.001), lower HDL-C (β = -20.8 [95% CI, -40.0 to -1.6]; P=0.034), and presence of hypertension (odds ratio, 1.2 [95% CI, 1.1-1.4]; P=0.0088), and the latter two were independent of BMI. Findings for diabetes mellitus were mixed and attenuated by adjusting for BMI. Metabolic syndrome was associated with MBH/AMY T2 signal ratios (odds ratio, 1.3 [95% CI, 1.1-1.6]; P<0.001). Model results were almost uniformly confirmed by the positive control ratios, whereas negative control ratios that did not test the MBH were unrelated to any outcomes (all P≥0.05). T2 signal ratios were not associated with prevalent coronary heart disease (all P>0.05), but confidence intervals were wide. Self-reported percentages of macronutrient intake were not consistently related to future T2 signal ratios. Conclusions Using a well-established study of cardiovascular disease development, we found evidence linking hypothalamic gliosis to multiple cardiovascular disease risk factors, even independent of adiposity. Our results highlight the need to consider neurologic mechanisms to understand and improve cardiometabolic health.
Collapse
Affiliation(s)
- Justin Lo
- School of Medicine, University of Washington, Seattle, WA
| | - Susan J Melhorn
- Department of Medicine, University of Washington, Seattle, WA
| | - Sarah Kee
- Department of Medicine, University of Washington, Seattle, WA
| | - Kelsey LW Olerich
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, University of Washington, Seattle, WA
| | - Alyssa Huang
- Department of Pediatrics, University of Washington, Seattle, WA
| | - Dabin Yeum
- Department of Medicine, University of Washington, Seattle, WA
| | - Alexa Beiser
- School of Public Health, Boston University, Boston, MA
| | - Sudha Seshadri
- Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Charles De Carli
- Department of Neurology, University of California, Davis, Davis, CA
| | - Ellen A Schur
- Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
4
|
Wei B, Cheng G, Bi Q, Lu C, Sun Q, Li L, Chen N, Hu M, Lu H, Xu X, Mao G, Wan S, Hu Z, Gu Y, Zheng J, Zhao L, Shen XZ, Liu X, Shi P. Microglia in the hypothalamic paraventricular nucleus sense hemodynamic disturbance and promote sympathetic excitation in hypertension. Immunity 2024; 57:2030-2042.e8. [PMID: 39116878 DOI: 10.1016/j.immuni.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 04/22/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
Hypertension is usually accompanied by elevated sympathetic tonicity, but how sympathetic hyperactivity is triggered is not clear. Recent advances revealed that microglia-centered neuroinflammation contributes to sympathetic excitation in hypertension. In this study, we performed a temporospatial analysis of microglia at both morphological and transcriptomic levels and found that microglia in the hypothalamic paraventricular nucleus (PVN), a sympathetic center, were early responders to hypertensive challenges. Vasculature analyses revealed that the PVN was characterized by high capillary density, thin vessel diameter, and complex vascular topology relative to other brain regions. As such, the PVN was susceptible to the penetration of ATP released from the vasculature in response to hemodynamic disturbance after blood pressure increase. Mechanistically, ATP ligation to microglial P2Y12 receptor was responsible for microglial inflammatory activation and the eventual sympathetic overflow. Together, these findings identified a distinct vasculature pattern rendering vulnerability of PVN pre-sympathetic neurons to hypertension-associated microglia-mediated inflammatory insults.
Collapse
Affiliation(s)
- Bo Wei
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Guo Cheng
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Qianqian Bi
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Cheng Lu
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Qihang Sun
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Li Li
- Department of Pharmacy, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China
| | - Ningting Chen
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Miner Hu
- Department of Cardiology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China
| | - Haoran Lu
- Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Haining 314400, China
| | - Xuancheng Xu
- Zhejiang Chinese Medical University, Hangzhou 310013, China; Department of Neurology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China
| | - Genxiang Mao
- Zhejiang Provincial Key Lab of Geriatrics, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shu Wan
- Brain Center, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China
| | - Zhechun Hu
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310058, China; Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Gu
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310058, China; Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaxin Zheng
- Key Laboratory for Biomedical Engineering of Ministrey of Education, Collage of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310013, China
| | - Li Zhao
- Key Laboratory for Biomedical Engineering of Ministrey of Education, Collage of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310013, China
| | - Xiao Z Shen
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Brain Center, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China; Department of Physiology, Zhejiang University School of Medicine, Hangzhou 310058, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310013, China
| | - Xiaoli Liu
- Department of Neurology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China.
| | - Peng Shi
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310013, China.
| |
Collapse
|
5
|
Ryan MJ, Clemmer JS, Mathew RO, Faulkner JL, Taylor EB, Abais-Battad JM, Hollis F, Sullivan JC. Revisiting sex as a biological variable in hypertension research. J Clin Invest 2024; 134:e180078. [PMID: 39225093 PMCID: PMC11364402 DOI: 10.1172/jci180078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Half of adults in the United States have hypertension as defined by clinical practice guidelines. Interestingly, women are generally more likely to be aware of their hypertension and have their blood pressure controlled with treatment compared with men, yet hypertension-related mortality is greater in women. This may reflect the fact that the female sex remains underrepresented in clinical and basic science studies investigating the effectiveness of therapies and the mechanisms controlling blood pressure. This Review provides an overview of the impact of the way hypertension research has explored sex as a biological variable (SABV). Emphasis is placed on epidemiological studies, hypertension clinical trials, the genetics of hypertension, sex differences in immunology and gut microbiota in hypertension, and the effect of sex on the central control of blood pressure. The goal is to offer historical perspective on SABV in hypertension, highlight recent studies that include SABV, and identify key gaps in SABV inclusion and questions that remain in the field. Through continued awareness campaigns and engagement/education at the level of funding agencies, individual investigators, and in the editorial peer review system, investigation of SABV in the field of hypertension research will ultimately lead to improved clinical outcomes.
Collapse
Affiliation(s)
- Michael J. Ryan
- Columbia VA Health Care System, Columbia, South Carolina, USA
- University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - John S. Clemmer
- University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Roy O. Mathew
- Loma Linda VA Health Care System, Loma Linda, California, USA
| | | | - Erin B. Taylor
- University of Mississippi Medical Center, Jackson, Mississippi, USA
| | | | - Fiona Hollis
- University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | | |
Collapse
|
6
|
Li J, Peng C, He K, Wang Y, Lai X. The central mechanisms of electroacupuncture at LR3 in the treatment of spontaneous hypertension: a PET and mRNA transcriptome study. Front Cardiovasc Med 2024; 11:1358426. [PMID: 39234603 PMCID: PMC11371727 DOI: 10.3389/fcvm.2024.1358426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
Objective To reveal the efficacy and potential mechanisms of electroacupuncture (EA) in treating hypertension. Methods Male spontaneously hypertensive rats (SHRs) were randomly assigned to the SHR group, EA group, and Sham-EA group, with Wistar-Kyoto rats (WKY) as the normal control group. SHRs in the EA group received electroacupuncture at the bilateral Taichong (LR3) acupoints for 7 consecutive days. Evaluation of systolic blood pressure (SBP), diastolic blood pressure (DBP), mean arterial pressure (MAP), and heart rate (HR) was conducted. Positron emission tomography-computed tomography (PET-CT) was employed to explore the active brain regions associated with acupuncture-induced blood pressure reduction. Furthermore, mRNA expression profiling was analyzed in the active brain regions to identify differentially expressed genes, and quantitative polymerase chain reaction (qPCR) was used to validate the mRNA expression of differentially expressed genes in the active brain region. Results EA reduced elevated SBP, DBP, MAP and HR in SHR. PET-CT revealed that EA decreased glucose metabolism in the hypothalamus. Genomic analysis suggested that, compared to the SHR group, the differentially expressed genes in the hypothalamus of the EA group included Nr4a1, Sirt1, Trh, GPR88, Cck, and Th. EA downregulated the mRNA expression of Th, Trh, Gpr88, and Nr4a1, while upregulating the expression of Sirt1 and Cck at the mRNA level. Conclusion EA may exert a unique antihypertensive effect in the hypothalamus of SHR, involving the modulation of sympathetic nerve activity, neuroinflammation, and oxidative stress response.
Collapse
Affiliation(s)
- Jing Li
- Integrative Cancer Centre, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Postdoctoral Research Station, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Clinical School of Acupuncture and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Chong Peng
- Postdoctoral Research Station, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Hepatobiliary Disease, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Kejie He
- Department of Acupuncture and Rehabilitation, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yumei Wang
- Department of Rehabilitation, Shenzhen Bao'an Traditional Chinese Medicine Hospital Group, Shenzhen, Guangdong, China
| | - Xinsheng Lai
- Clinical School of Acupuncture and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Schwalbe DC, Stornetta DS, Abraham-Fan RJ, Souza GMPR, Jalil M, Crook ME, Campbell JN, Abbott SBG. Molecular Organization of Autonomic, Respiratory, and Spinally-Projecting Neurons in the Mouse Ventrolateral Medulla. J Neurosci 2024; 44:e2211232024. [PMID: 38918066 PMCID: PMC11293450 DOI: 10.1523/jneurosci.2211-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024] Open
Abstract
The ventrolateral medulla (VLM) is a crucial region in the brain for visceral and somatic control, serving as a significant source of synaptic input to the spinal cord. Experimental studies have shown that gene expression in individual VLM neurons is predictive of their function. However, the molecular and cellular organization of the VLM has remained uncertain. This study aimed to create a comprehensive dataset of VLM cells using single-cell RNA sequencing in male and female mice. The dataset was enriched with targeted sequencing of spinally-projecting and adrenergic/noradrenergic VLM neurons. Based on differentially expressed genes, the resulting dataset of 114,805 VLM cells identifies 23 subtypes of neurons, excluding those in the inferior olive, and five subtypes of astrocytes. Spinally-projecting neurons were found to be abundant in seven subtypes of neurons, which were validated through in situ hybridization. These subtypes included adrenergic/noradrenergic neurons, serotonergic neurons, and neurons expressing gene markers associated with premotor neurons in the ventromedial medulla. Further analysis of adrenergic/noradrenergic neurons and serotonergic neurons identified nine and six subtypes, respectively, within each class of monoaminergic neurons. Marker genes that identify the neural network responsible for breathing were concentrated in two subtypes of neurons, delineated from each other by markers for excitatory and inhibitory neurons. These datasets are available for public download and for analysis with a user-friendly interface. Collectively, this study provides a fine-scale molecular identification of cells in the VLM, forming the foundation for a better understanding of the VLM's role in vital functions and motor control.
Collapse
Affiliation(s)
- Dana C Schwalbe
- Departments of Biology, University of Virginia, Charlottesville, Virginia 22904
| | | | | | | | - Maira Jalil
- Departments of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Maisie E Crook
- Departments of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - John N Campbell
- Departments of Biology, University of Virginia, Charlottesville, Virginia 22904
| | | |
Collapse
|
8
|
Shen B, Yang L, Jia X, Kong D, Jing L, Gao Y, Gao S, Chen R, Chen F, Zhao C, Li Y, Tan R, Zhao X. Contribution of platelets to disruption of the blood-brain barrier during arterial baroreflex dysfunction. Microvasc Res 2024; 154:104681. [PMID: 38493885 DOI: 10.1016/j.mvr.2024.104681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Arterial baroreflex dysfunction, like many other central nervous system disorders, involves disruption of the blood-brain barrier, but what causes such disruption in ABR dysfunction is unclear. Here we explored the potential role of platelets in this disruption. METHODS ABR dysfunction was induced in rats using sinoaortic denervation, and the effects on integrity of the blood-brain barrier were explored based on leakage of Evans blue or FITC-dextran, while the effects on expression of CD40L in platelets and of key proteins in microvascular endothelial cells were explored using immunohistochemistry, western blotting and enzyme-linked immunosorbent assay. Similar experiments were carried out in rat brain microvascular endothelial cell line, which we exposed to platelets taken from rats with ABR dysfunction. RESULTS Sinoaortic denervation permeabilized the blood-brain barrier and downregulated zonula occludens-1 and occludin in rat brain, while upregulating expression of CD40L on the surface of platelets and stimulating platelet aggregation. Similar effects of permeabilization and downregulation were observed in healthy rats that received platelets from animals with ABR dysfunction, and in rat brain microvascular endothelial cells, but only in the presence of lipopolysaccharide. These effects were associated with activation of NF-κB signaling and upregulation of matrix metalloprotease-9. These effects of platelets from animals with ABR dysfunction were partially blocked by neutralizing antibody against CD40L or the platelet inhibitor clopidogrel. CONCLUSION During ABR dysfunction, platelets may disrupt the blood-brain barrier when CD40L on their surface activates NF-kB signaling within cerebral microvascular endothelial cells, leading to upregulation of matrix metalloprotease-9. Our findings imply that targeting CD40L may be effective against cerebral diseases involving ABR dysfunction.
Collapse
Affiliation(s)
- Bowen Shen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Lili Yang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Xiaoli Jia
- Department of Pharmacy, Liaocheng People's Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Liao'cheng 252000, China
| | - Deping Kong
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Lei Jing
- Department of Pharmacy, Dongping People's Hospital, Tai'an 271500, China
| | - Yongfeng Gao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Shan Gao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Ruimin Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Fengbao Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Chunyu Zhao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Yue Li
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Rui Tan
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China.
| | - Xiaomin Zhao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China.
| |
Collapse
|
9
|
Boomer SH, Liu X, Zheng H. Effects of regulator of G protein signaling 2 (RGS2) overexpression in the paraventricular nucleus on blood pressure in rats with angiotensin II-induced hypertension. Front Physiol 2024; 15:1401768. [PMID: 38974519 PMCID: PMC11224644 DOI: 10.3389/fphys.2024.1401768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 05/09/2024] [Indexed: 07/09/2024] Open
Abstract
The hypothalamic paraventricular nucleus (PVN) regulates sympathetic activity and blood pressure. The regulator of G protein signaling 2 (RGS2) is a negative G protein regulator, which selectively regulates G⍺q signaling, a potential cause of hypertension. This study aimed to examine angiotensin II (ANG II)-G protein-RGS2 signaling on the central mechanisms of blood pressure control, sympathetic activation, and kidney function. The Sprague Dawley rats were infused with ANG II (200 ng/kg/min) via osmotic mini pump to induce hypertension. Adenovirus (AV) vectors encoding RGS2 was transfected into the PVN in vivo. By radio telemetry measurements, we found AV-RGS2 transfection to the PVN significantly attenuated the increase of mean arterial pressure in ANG II infusion rats from days 2-7 of the 2-week experiment (Day 7: ANG II + AV-RGS2 141.3 ± 10.0 mmHg vs. ANG II 166.9 ± 9.3 mmHg, p < 0.05). AV-RGS2 transfection significantly reduced the serum norepinephrine level and acute volume reflex and increased daily urine volume and sodium excretion in ANG II-infused hypertensive rats. AV-RGS2 transfection significantly reduced G⍺q and PKC protein expressions within the PVN in ANG II infusion rats. In cultured mouse hypothalamic cells, real-time PCR study showed ANG II treatment increased mRNA expression of G⍺q, G⍺s, and RGS2, and AV-RGS2 treatment decreased ANG II-induced mRNA expression of G⍺q and G⍺s. Using confocal imagery, we found that AV-RGS2 attenuated the increase of calcium influx in ANG II-treated cells. Our results suggest that central overexpression of RGS2 in the PVN attenuated the increase of blood pressure and sympathetic outflow, and improves kidney excretory function in hypertensive rats. This may be via the alteration of ANG II-G-protein-RGS2 signaling in the central nervous system.
Collapse
Affiliation(s)
| | | | - Hong Zheng
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| |
Collapse
|
10
|
Li KH, Lin JM, Luo SQ, Li MY, Yang YY, Li MM, Xia PY, Su JZ. Afferent Renal Denervation Attenuates Sympathetic Overactivation From the Paraventricular Nucleus in Spontaneously Hypertensive Rats. Am J Hypertens 2024; 37:477-484. [PMID: 38459938 DOI: 10.1093/ajh/hpae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/06/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024] Open
Abstract
BACKGROUND The effectiveness of renal denervation (RDN) in reducing blood pressure and systemic sympathetic activity in hypertensive patients has been established. However, the underlying central mechanism remains unknown. This study aimed to investigate the role of RDN in regulating cardiovascular function via the central renin-angiotensin system (RAS) pathway. METHODS Ten-week-old spontaneously hypertensive rats (SHR) were subjected to selective afferent renal denervation (ADN) using capsaicin solution. We hypothesized that ADN would effectively reduce blood pressure and rebalance the RAS component of the paraventricular nucleus (PVN) in SHR. RESULTS The experimental results show that the ADN group exhibited significantly lower blood pressure, reduced systemic sympathetic activity, decreased chronic neuronal activation marker C-FOS expression in the PVN, and improved arterial baroreflex function, compared with the Sham group. Furthermore, ACE and AT1 protein expression was reduced while ACE2 and MAS protein expression was increased in the PVN of SHR after ADN. CONCLUSIONS These findings suggest that RDN may exert these beneficial effects through modulating the central RAS pathway.
Collapse
Affiliation(s)
- Kun-Hui Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
- Department of Rehabilitation Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Jie-Min Lin
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
- Department of Rehabilitation Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Si-Qi Luo
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
- Department of Rehabilitation Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Min-Yan Li
- Department of Rehabilitation and Health, Fujian Vocational College of Bioengineering, Fuzhou, China
| | - Yi-Yong Yang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
- Department of Rehabilitation Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Meng-Meng Li
- Department of Cardiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Pan-Yan Xia
- Department of Rehabilitation Medicine, The School of Health, Fujian Medical University, Fuzhou, China
| | - Jin-Zi Su
- Department of Cardiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
11
|
Rahmouni K. Neural Circuits Underlying Reciprocal Cardiometabolic Crosstalk: 2023 Arthur C. Corcoran Memorial Lecture. Hypertension 2024; 81:1233-1243. [PMID: 38533662 PMCID: PMC11096079 DOI: 10.1161/hypertensionaha.124.22066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
The interplay of various body systems, encompassing those that govern cardiovascular and metabolic functions, has evolved alongside the development of multicellular organisms. This evolutionary process is essential for the coordination and maintenance of homeostasis and overall health by facilitating the adaptation of the organism to internal and external cues. Disruption of these complex interactions contributes to the development and progression of pathologies that involve multiple organs. Obesity-associated cardiovascular risks, such as hypertension, highlight the significant influence that metabolic processes exert on the cardiovascular system. This cardiometabolic communication is reciprocal, as indicated by substantial evidence pointing to the ability of the cardiovascular system to affect metabolic processes, with pathophysiological implications in disease conditions. In this review, I outline the bidirectional nature of the cardiometabolic interaction, with special emphasis on the impact that metabolic organs have on the cardiovascular system. I also discuss the contribution of the neural circuits and autonomic nervous system in mediating the crosstalk between cardiovascular and metabolic functions in health and disease, along with the molecular mechanisms involved.
Collapse
Affiliation(s)
- Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Veterans Affairs Health Care System, Iowa City, Iowa
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Obesity Research and Education Initiative, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
12
|
Karlen-Amarante M, Glovak ZT, Huff A, Oliveira LM, Ramirez JM. Postinspiratory and preBötzinger complexes contribute to respiratory-sympathetic coupling in mice before and after chronic intermittent hypoxia. Front Neurosci 2024; 18:1386737. [PMID: 38774786 PMCID: PMC11107097 DOI: 10.3389/fnins.2024.1386737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/16/2024] [Indexed: 05/24/2024] Open
Abstract
The sympathetic nervous system modulates arterial blood pressure. Individuals with obstructive sleep apnea (OSA) experience numerous nightly hypoxic episodes and exhibit elevated sympathetic activity to the cardiovascular system leading to hypertension. This suggests that OSA disrupts normal respiratory-sympathetic coupling. This study investigates the role of the postinspiratory complex (PiCo) and preBötzinger complex (preBötC) in respiratory-sympathetic coupling under control conditions and following exposure to chronic intermittent hypoxia (CIH) for 21 days (5% O2-80 bouts/day). The surface of the ventral brainstem was exposed in urethane (1.5 g/kg) anesthetized, spontaneously breathing adult mice. Cholinergic (ChAT), glutamatergic (Vglut2), and neurons that co-express ChAT and Vglut2 at PiCo, as well as Dbx1 and Vglut2 neurons at preBötC, were optogenetically stimulated while recording activity from the diaphragm (DIA), vagus nerve (cVN), and cervical sympathetic nerve (cSN). Following CIH exposure, baseline cSN activity increased, breathing frequency increased, and expiratory time decreased. In control mice, stimulating PiCo specific cholinergic-glutamatergic neurons caused a sympathetic burst during all phases of the respiratory cycle, whereas optogenetic activation of cholinergic-glutamatergic PiCo neurons in CIH mice increased sympathetic activity only during postinspiration and late expiration. Stimulation of glutamatergic PiCo neurons increased cSN activity during the postinspiratory phase in control and CIH mice. Optogenetic stimulation of ChAT containing neurons in the PiCo area did not affect sympathetic activity under control or CIH conditions. Stimulating Dbx1 or Vglut2 neurons in preBötC evoked an inspiration and a concomitant cSN burst under control and CIH conditions. Taken together, these results suggest that PiCo and preBötC contribute to respiratory-sympathetic coupling, which is altered by CIH, and may contribute to the hypertension observed in patients with OSA.
Collapse
Affiliation(s)
- Marlusa Karlen-Amarante
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Zachary T. Glovak
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Alyssa Huff
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Luiz M. Oliveira
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Neurological Surgery, University of Washington, Seattle, WA, United States
| |
Collapse
|
13
|
Hu M, Xu F, Liu S, Yao Y, Xia Q, Zhu C, Zhang X, Tang H, Qaiser Z, Liu S, Tang Y. Aging pattern of the brainstem based on volumetric measurement and optimized surface shape analysis. Brain Imaging Behav 2024; 18:396-411. [PMID: 38155336 DOI: 10.1007/s11682-023-00840-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2023] [Indexed: 12/30/2023]
Abstract
The brainstem, a small and crucial structure, is connected to the cerebrum, spinal cord, and cerebellum, playing a vital role in regulating autonomic functions, transmitting motor and sensory information, and modulating cognitive processes, emotions, and consciousness. While previous research has indicated that changes in brainstem anatomy can serve as a biomarker for aging and neurodegenerative diseases, the structural changes that occur in the brainstem during normal aging remain unclear. This study aimed to examine the age- and sex-related differences in the global and local structural measures of the brainstem in 187 healthy adults (ranging in age from 18 to 70 years) using structural magnetic resonance imaging. The findings showed a significant negative age effect on the volume of the two major components of the brainstem: the medulla oblongata and midbrain. The shape analysis revealed that atrophy primarily occurs in specific structures, such as the pyramid, cerebral peduncle, superior and inferior colliculi. Surface area and shape analysis showed a trend of flattening in the aging brainstem. There were no significant differences between the sexes or sex-by-age interactions in brainstem structural measures. These findings provide a systematic description of age associations with brainstem structures in healthy adults and may provide a reference for future research on brain aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Minqi Hu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Feifei Xu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Shizhou Liu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Yuan Yao
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Qing Xia
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Caiting Zhu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Xinwen Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Haiyan Tang
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Zubair Qaiser
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Shuwei Liu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Yuchun Tang
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China.
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
14
|
Stocker SD, Ferreira CB, Souza GM, Abbott SB. Brain Pathways in Blood Pressure Regulation. Hypertension 2024; 81:383-386. [PMID: 38193317 PMCID: PMC11003736 DOI: 10.1161/hypertensionaha.123.21723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Affiliation(s)
- Sean D. Stocker
- Department of Neurobiology, University of Pittsburgh School of Medicine
| | | | | | | |
Collapse
|
15
|
Moss A, Kuttippurathu L, Srivastava A, Schwaber JS, Vadigepalli R. Dynamic dysregulation of transcriptomic networks in brainstem autonomic nuclei during hypertension development in the female spontaneously hypertensive rat. Physiol Genomics 2024; 56:283-300. [PMID: 38145287 PMCID: PMC11283910 DOI: 10.1152/physiolgenomics.00073.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023] Open
Abstract
Neurogenic hypertension stems from an imbalance in autonomic function that shifts the central cardiovascular control circuits toward a state of dysfunction. Using the female spontaneously hypertensive rat and the normotensive Wistar-Kyoto rat model, we compared the transcriptomic changes in three autonomic nuclei in the brainstem, nucleus of the solitary tract (NTS), caudal ventrolateral medulla, and rostral ventrolateral medulla (RVLM) in a time series at 8, 10, 12, 16, and 24 wk of age, spanning the prehypertensive stage through extended chronic hypertension. RNA-sequencing data were analyzed using an unbiased, dynamic pattern-based approach that uncovered dominant and several subtle differential gene regulatory signatures. Our results showed a persistent dysregulation across all three autonomic nuclei regardless of the stage of hypertension development as well as a cascade of transient dysregulation beginning in the RVLM at the prehypertensive stage that shifts toward the NTS at the hypertension onset. Genes that were persistently dysregulated were heavily enriched for immunological processes such as antigen processing and presentation, the adaptive immune response, and the complement system. Genes with transient dysregulation were also largely region-specific and were annotated for processes that influence neuronal excitability such as synaptic vesicle release, neurotransmitter transport, and an array of neuropeptides and ion channels. Our results demonstrate that neurogenic hypertension is characterized by brainstem region-specific transcriptomic changes that are highly dynamic with significant gene regulatory changes occurring at the hypertension onset as a key time window for dysregulation of homeostatic processes across the autonomic control circuits.NEW & NOTEWORTHY Hypertension is a major disease and is the primary risk factor for cardiovascular complications and stroke. The gene expression changes in the central nervous system circuits driving hypertension are understudied. Here, we show that coordinated and region-specific gene expression changes occur in the brainstem autonomic circuits over time during the development of a high blood pressure phenotype in a rat model of human essential hypertension.
Collapse
Affiliation(s)
- Alison Moss
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Lakshmi Kuttippurathu
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Ankita Srivastava
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - James S Schwaber
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| |
Collapse
|
16
|
Sotozawa M, Kinguchi S, Wakui H, Azushima K, Funakoshi K, Nakajima W, Miyazaki T, Takahashi T, Tamura K. Enhancement of angiotensin II type 1 receptor-associated protein in the paraventricular nucleus suppresses angiotensin II-dependent hypertension. Hypertens Res 2024; 47:67-77. [PMID: 37884662 DOI: 10.1038/s41440-023-01480-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/29/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023]
Abstract
The renin-angiotensin system in the brain plays a pivotal role in modulating sympathetic nerve activity and contributes to the pathogenesis of hypertension. Angiotensin II (Ang II) type 1 receptor (AT1R)-associated protein (ATRAP) promotes internalization of AT1R while suppressing pathological overactivation of AT1R signaling. However, the pathophysiological function of ATRAP in the brain remains unknown. Therefore, this study aims to investigate whether ATRAP in the paraventricular nucleus (PVN) is involved in neurogenic hypertension pathogenesis in Ang II-infused rats. The ATRAP/AT1R ratio, which serves as an indicator of tissue AT1R hyperactivity, tended to decrease within the PVN in the Ang II group than in the vehicle group. This suggests an Ang II-induced hyperactivation of the AT1R signaling pathway in the PVN. Lentiviral vectors were generated to stimulate ATRAP expression. At 6 weeks of age, rats were microinjected with LV-Venus (Venus-expressing lentivirus) or LV-ATRAP (Venus-ATRAP-expressing lentivirus). The rats were then randomly divided into four groups: (1) Vehicle/LV-Venus, (2) Vehicle/LV-ATRAP, (3) Ang II/LV-Venus, and (4) Ang II/LV-ATRAP. Two weeks after microinjection, vehicle or Ang II was administered systemically for 2 weeks. In the Ang II/LV-ATRAP group, systolic blood pressure at 1 and 2 weeks following administration was significantly lower than that in the Ang II/LV-Venus group. Furthermore, urinary adrenaline levels tended to decrease in the Ang II/LV-ATRAP group than in the Ang II/LV-Venus group. These findings suggest that enhanced ATRAP expression in the PVN suppresses Ang II-induced hypertension, potentially by suppressing hyperactivation of the tissue AT1R signaling pathway and, subsequently, sympathetic nerve activity.
Collapse
Affiliation(s)
- Mari Sotozawa
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sho Kinguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Kengo Funakoshi
- Department of Neuroanatomy, Yokohama City University School of Medicine, Yokohama, Japan
| | - Waki Nakajima
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomoyuki Miyazaki
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takuya Takahashi
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
17
|
Dellacqua LO, Gomes PM, Batista JS, Michelini LC, Antunes VR. Exercise-induced neuroplasticity in autonomic nuclei restores the cardiac vagal tone and baroreflex dysfunction in aged hypertensive rats. J Appl Physiol (1985) 2024; 136:189-198. [PMID: 38059293 DOI: 10.1152/japplphysiol.00433.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023] Open
Abstract
Aging is accompanied by considerable deterioration of homeostatic systems, such as autonomic imbalance characterized by heightened sympathetic activity, lower parasympathetic tone, and depressed heart rate (HR) variability, which are aggravated by hypertension. Here, we hypothesized that these age-related deficits in aged hypertensive rats can be ameliorated by exercise training, with benefits to the cardiovascular system. Therefore, male 22-mo-old spontaneously hypertensive rats (SHRs) and age-matched Wistar Kyoto (WKY) submitted to moderate-intensity exercise training (T) or kept sedentary (S) for 8 wk were evaluated for hemodynamic/autonomic parameters, baroreflex sensitivity, cardiac sympathetic/parasympathetic tone and analysis of dopamine β-hydroxylase (DBH+) and oxytocin (OT+) pathways of autonomic brain nuclei. Aged SHR-S versus WKY-S exhibited elevated mean arterial pressure (MAP: +51%) and HR (+20%), augmented pressure/HR variability, no cardiac vagal tone, and depressed reflex control of the heart (HR range, -28%; gain, -49%). SHR-T exhibited a lower resting HR, a partial reduction in the MAP (-14%), in the pressure/HR variabilities, and restored parasympathetic modulation, with improvement of baroreceptor reflex control when compared with SHR-S. Exercise training increased the ascending DBH+ projections conveying peripheral information to the paraventricular nucleus of hypothalamus (PVN), augmented the expression of OT+ neurons, and reduced the density of DBH+ neurons in the rostral ventrolateral medulla (RVLM) of SHR-T. Data indicate that exercise training induces beneficial neuroplasticity in brain autonomic circuitry, and it is highly effective to restore the parasympathetic tone, and attenuation of age-related autonomic imbalance and baroreflex dysfunction, thus conferring long-term benefits for cardiovascular control in aged hypertensive individuals.NEW & NOTEWORTHY Exercise training reduces high blood pressure and cardiovascular autonomic modulation in aged hypertensive rats. The dysfunction in the baroreflex sensitivity and impaired parasympathetic tone to the heart of aged hypertensive rats are restored by exercise training. Exercise induces beneficial neuroplasticity in the brain nuclei involved with autonomic control of cardiovascular function of aged hypertensive rats.
Collapse
Affiliation(s)
- Lais Oliveira Dellacqua
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Paula Magalhães Gomes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Julia Santos Batista
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Lisete Compagno Michelini
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Vagner Roberto Antunes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
18
|
Lohman T, Sible I, Kapoor A, Engstrom AC, Alitin JP, Gaubert A, Rodgers KE, Bradford D, Mather M, Han SD, Thayer JF, Nation DA. Blood pressure variability, central autonomic network dysfunction and cerebral small vessel disease in APOE4 carriers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.13.23299556. [PMID: 38168394 PMCID: PMC10760290 DOI: 10.1101/2023.12.13.23299556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Background Increased blood pressure variability (BPV) is a risk factor for cerebral small vessel disease (CSVD) and neurodegeneration, independent of age and average blood pressure, particularly in apolipoprotein E4 (APOE4) carriers. However, it remains uncertain whether BPV elevation is a cause or a consequence of vascular brain injury, or to what degree injury to the central autonomic network (CAN) may contribute to BPV-associated risk in APOE4 carriers. Methods Independently living older adults (n=70) with no history of stroke or dementia were recruited from the community and underwent 5 minutes of resting beat-to-beat blood pressure monitoring, genetic testing, and brain MRI. Resting BPV, APOE genotype, CSVD burden on brain MRI, and resting state CAN connectivity by fMRI were analyzed. Causal mediation and moderation analysis evaluated BPV and CAN effects on CSVD in APOE4 carriers (n=37) and non-carriers (n=33). Results Higher BPV was associated with the presence and extent of CSVD in APOE4 carriers, but not non-carriers, independent of CAN connectivity (B= 18.92, P= .02), and CAN connectivity did not mediate the relationship between BPV and CSVD. In APOE4 carriers, CAN connectivity moderated the relationship between BPV and CSVD, whereby BPV effects on CSVD were greater in those with lower CAN connectivity (B= 36.43, P= .02). Conclusions Older APOE4 carriers with higher beat-to-beat BPV exhibit more extensive CSVD, independent of average blood pressure, and the strength of CAN connectivity does not mediate these effects. Findings suggest increased BPV is more likely a cause, not a consequence, of CSVD. BPV is more strongly associated with CSVD in APOE4 carriers with lower rsCAN connectivity, suggesting CAN dysfunction and BPV elevation may have synergistic effects on CSVD. Further studies are warranted to understand the interplay between BPV and CAN function in APOE4 carriers.
Collapse
Affiliation(s)
- Trevor Lohman
- University of Southern California Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - Isabel Sible
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Arunima Kapoor
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Allison C Engstrom
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - John Paul Alitin
- University of Southern California Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - Aimee Gaubert
- University of Southern California Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - Kathleen E Rodgers
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - David Bradford
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Mara Mather
- University of Southern California Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - S Duke Han
- University of Southern California Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - Julian F Thayer
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Daniel A Nation
- University of Southern California Leonard Davis School of Gerontology, Los Angeles, CA, USA
- University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
19
|
Clayton TL. Obesity and hypertension: Obesity medicine association (OMA) clinical practice statement (CPS) 2023. OBESITY PILLARS 2023; 8:100083. [PMID: 38125655 PMCID: PMC10728712 DOI: 10.1016/j.obpill.2023.100083] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 08/06/2023] [Indexed: 12/23/2023]
Abstract
Background This Obesity Medicine Association (OMA) Clinical Practice Statement (CPS) provides an overview of the mechanisms and treatment of obesity and hypertension. Methods The scientific support for this CPS is based upon published citations, clinical perspectives of OMA authors, and peer review by the Obesity Medicine Association leadership. Results Mechanisms contributing to obesity-related hypertension include unhealthful nutrition, physical inactivity, insulin resistance, increased sympathetic nervous system activity, renal dysfunction, vascular dysfunction, heart dysfunction, increased pancreatic insulin secretion, sleep apnea, and psychosocial stress. Adiposopathic factors that may contribute to hypertension include increased release of free fatty acids, increased leptin, decreased adiponectin, increased renin-angiotensin-aldosterone system activation, increased 11 beta-hydroxysteroid dehydrogenase type 1, reduced nitric oxide activity, and increased inflammation. Conclusions Increase in body fat is the most common cause of hypertension. Among patients with obesity and hypertension, weight reduction via healthful nutrition, physical activity, behavior modification, bariatric surgery, and anti-obesity medications mostly decrease blood pressure, with the greatest degree of weight reduction generally correlated with the greatest degree of blood pressure reduction.
Collapse
Affiliation(s)
- Tiffany Lowe Clayton
- Diplomate of American Board of Obesity Medicine, WakeMed Bariatric Surgery and Medical Weight Loss USA
- Campbell University School of Osteopathic Medicine, Buies Creek, NC 27546, Levine Hall Room 170 USA
| |
Collapse
|
20
|
Pace SA, Myers B. Hindbrain Adrenergic/Noradrenergic Control of Integrated Endocrine and Autonomic Stress Responses. Endocrinology 2023; 165:bqad178. [PMID: 38015813 DOI: 10.1210/endocr/bqad178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/07/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Hindbrain adrenergic/noradrenergic nuclei facilitate endocrine and autonomic responses to physical and psychological challenges. Neurons that synthesize adrenaline and noradrenaline target hypothalamic structures to modulate endocrine responses while descending spinal projections regulate sympathetic function. Furthermore, these neurons respond to diverse stress-related metabolic, autonomic, and psychosocial challenges. Accordingly, adrenergic and noradrenergic nuclei are integrative hubs that promote physiological adaptation to maintain homeostasis. However, the precise mechanisms through which adrenaline- and noradrenaline-synthesizing neurons sense interoceptive and exteroceptive cues to coordinate physiological responses have yet to be fully elucidated. Additionally, the regulatory role of these cells in the context of chronic stress has received limited attention. This mini-review consolidates reports from preclinical rodent studies on the organization and function of brainstem adrenaline and noradrenaline cells to provide a framework for how these nuclei coordinate endocrine and autonomic physiology. This includes identification of hindbrain adrenaline- and noradrenaline-producing cell groups and their role in stress responding through neurosecretory and autonomic engagement. Although temporally and mechanistically distinct, the endocrine and autonomic stress axes are complementary and interconnected. Therefore, the interplay between brainstem adrenergic/noradrenergic nuclei and peripheral physiological systems is necessary for integrated stress responses and organismal survival.
Collapse
Affiliation(s)
- Sebastian A Pace
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Brent Myers
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
21
|
Rodrigues AF, Bader M. The contribution of the AT1 receptor to erythropoiesis. Biochem Pharmacol 2023; 217:115805. [PMID: 37714274 DOI: 10.1016/j.bcp.2023.115805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
The renin-angiotensin system (RAS) comprises a broad set of functional peptides and receptors that play a role in cardiovascular homeostasis and contribute to cardiovascular pathologies. Angiotensin II (Ang II) is the most potent peptide hormone produced by the RAS due to its high abundance and its strong and pleiotropic impact on the cardiovascular system. Formation of Ang II takes place in the bloodstream and additionally in tissues in the so-called local RAS. Of the two Ang II receptors (AT1 and AT2) that Ang II binds to, AT1 is the most expressed throughout the mammalian body. AT1 expression is not restricted to cells of the cardiovascular system but in fact AT1 protein is found in nearly all organs, hence, Ang II takes part in several modulatory physiological processes one of which is erythropoiesis. In this review, we present multiple evidence supporting that Ang II modulates physiological and pathological erythropoiesis processes trough the AT1 receptor. Cumulative evidence indicates that Ang II by three distinct mechanisms influences erythropoiesis: 1) stimulation of renal erythropoietin synthesis; 2) direct action on bone marrow precursor cells; and 3) modulation of sympathetic nerve activity to the bone marrow. The text highlights clinical and preclinical evidence focusing on mechanistic studies using rodent models.
Collapse
Affiliation(s)
- André F Rodrigues
- Max Delbrück Center (MDC), Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany.
| | - Michael Bader
- Max Delbrück Center (MDC), Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany; Charité Universitätsmedizin Berlin, Berlin, Germany; Institute for Biology, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
22
|
Sun Q, Li L, Jin F, Liu Y, Yang B, Meng W, Zhang Z, Qi F. SARS-CoV-2 Spike Protein S1 Exposure Increases Susceptibility to Angiotensin II-Induced Hypertension in Rats by Promoting Central Neuroinflammation and Oxidative Stress. Neurochem Res 2023; 48:3016-3026. [PMID: 37269471 PMCID: PMC10239221 DOI: 10.1007/s11064-023-03949-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 06/05/2023]
Abstract
The SARS-CoV-2 spike S1 subunit (S1) can cross the blood-brain barrier and elicit neuroinflammatory response independent of viral infection. Here we examined whether S1 influences blood pressure (BP) and sensitizes the hypertensive response to angiotensin (ANG) II by enhancing neuroinflammation and oxidative stress in hypothalamic paraventricular nucleus (PVN), a key brain cardiovascular regulatory center. Rats received central S1 or vehicle (VEH) injection for 5 days. One week after injection, ANG II or saline (control) was subcutaneously delivered for 2 weeks. S1 injection induced greater increases in BP, PVN neuronal excitation and sympathetic drive in ANG II rats but had no effects in control rats. One week after S1 injection, mRNA for proinflammatory cytokines and oxidative stress marker were higher but mRNA of Nrf2, the master regulator of inducible antioxidant and anti-inflammatory responses, was lower in the PVN in S1-injected rats than in VEH-injected rats. Three weeks after S1 injection, mRNA for proinflammatory cytokines and oxidative stress marker, microglia activation and reactive oxygen species in the PVN were comparable between S1 and VEH treated control rats but were elevated in two groups of ANG II rats. Notably, ANG II-induced elevations in these parameters were exaggerated by S1. Interestingly, ANG II increased PVN Nrf2 mRNA in VEH-treated rats but not in S1-treated rats. These data suggest that S1 exposure has no effect on BP, but post-S1 exposure increases susceptibility to ANG II-induced hypertension by downregulating PVN Nrf2 to promote neuroinflammation and oxidative stress and augment sympathetic excitation.
Collapse
Affiliation(s)
- Qingmei Sun
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China
| | - Liang Li
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China
| | - Feihong Jin
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China
| | - Yu Liu
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China
| | - Bo Yang
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China
| | - Wanping Meng
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China
| | - Zibin Zhang
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China
| | - Feng Qi
- Department of Anesthesiology, Qilu Hospital of Shandong University, No.107 Wenhua Xi Road, Jinan, 250012, China.
| |
Collapse
|
23
|
Wafi AM. Nrf2 and autonomic dysregulation in chronic heart failure and hypertension. Front Physiol 2023; 14:1206527. [PMID: 37719456 PMCID: PMC10500196 DOI: 10.3389/fphys.2023.1206527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 08/14/2023] [Indexed: 09/19/2023] Open
Abstract
Redox imbalance plays essential role in the pathogenesis of cardiovascular diseases. Chronic heart failure (CHF) and hypertension are associated with central oxidative stress, which is partly mediated by the downregulation of antioxidant enzymes in the central autonomic neurons that regulate sympathetic outflow, resulting in sympathoexcitation. Antioxidant proteins are partially regulated by the transcriptional factor nuclear factor erythroid 2-related factor 2 (Nrf2). Downregulation of Nrf2 is key to disrupting central redox homeostasis and mediating sympathetic nerve activity in the setting of Chronic heart failure and hypertension. Nrf2, in turn, is regulated by various mechanisms, such as extracellular vesicle-enriched microRNAs derived from several cell types, including heart and skeletal muscle. In this review, we discuss the role of Nrf2 in regulating oxidative stress in the brain and its impact on sympathoexcitation in Chronic heart failure and hypertension. Importantly, we also discuss interorgan communication via extracellular vesicle pathways that mediate central redox imbalance through Nrf2 signaling.
Collapse
Affiliation(s)
- Ahmed M. Wafi
- Physiology Department, Faculty of Medicine, Jazan University, Jizan, Saudi Arabia
| |
Collapse
|
24
|
Jun S, Ou X, Shi L, Yu H, Deng T, Chen J, Nie X, Hao Y, Shi Y, Liu W, Tian Y, Wang S, Yuan F. Circuit-Specific Control of Blood Pressure by PNMT-Expressing Nucleus Tractus Solitarii Neurons. Neurosci Bull 2023; 39:1193-1209. [PMID: 36588135 PMCID: PMC10387028 DOI: 10.1007/s12264-022-01008-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/18/2022] [Indexed: 01/03/2023] Open
Abstract
The nucleus tractus solitarii (NTS) is one of the morphologically and functionally defined centers that engage in the autonomic regulation of cardiovascular activity. Phenotypically-characterized NTS neurons have been implicated in the differential regulation of blood pressure (BP). Here, we investigated whether phenylethanolamine N-methyltransferase (PNMT)-expressing NTS (NTSPNMT) neurons contribute to the control of BP. We demonstrate that photostimulation of NTSPNMT neurons has variable effects on BP. A depressor response was produced during optogenetic stimulation of NTSPNMT neurons projecting to the paraventricular nucleus of the hypothalamus, lateral parabrachial nucleus, and caudal ventrolateral medulla. Conversely, photostimulation of NTSPNMT neurons projecting to the rostral ventrolateral medulla produced a robust pressor response and bradycardia. In addition, genetic ablation of both NTSPNMT neurons and those projecting to the rostral ventrolateral medulla impaired the arterial baroreflex. Overall, we revealed the neuronal phenotype- and circuit-specific mechanisms underlying the contribution of NTSPNMT neurons to the regulation of BP.
Collapse
Affiliation(s)
- Shirui Jun
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xianhong Ou
- Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Luo Shi
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Hongxiao Yu
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Tianjiao Deng
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jinting Chen
- Core Facilities and Centers, Institute of Medicine and Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xiaojun Nie
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yinchao Hao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yishuo Shi
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Wei Liu
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yanming Tian
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Sheng Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China.
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, 050017, China.
| | - Fang Yuan
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China.
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, 050017, China.
| |
Collapse
|
25
|
Tong L, Chen G, Liu T, Wang L, Zhang H, Chen F, Zhang S, Du D. IFN-γ deficiency in the rostral ventrolateral medulla contributes to stress-induced hypertension by impairing microglial synaptic engulfment. J Hypertens 2023; 41:1323-1332. [PMID: 37260264 DOI: 10.1097/hjh.0000000000003470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
BACKGROUND Dysfunctional neurons and microglia in the rostral ventrolateral medulla (RVLM) have been implicated in the pathogenesis of stress-induced hypertension (SIH). Functional perturbation of microglial synaptic engulfment can induce aberrant brain circuit activity. IFN-γ is a pleiotropic cytokine that plays a role in regulating neuronal activity. However, existing research on the exploration of the effects of microglia on synapses in the RVLM is lacking, particularly on the function of IFN-γ in microglial synaptic engulfment involved in SIH. METHODS A SIH rat model was established by electric foot shocks combined with noise stimulation. The underlying mechanism of IFN-γ on synaptic density and microglial synaptic engulfment was investigated through in-vivo and in-vitro experiments involving gain of function, immunofluorescence, quantitative real-time PCR, western blot, and morphometric analysis. Furthermore, the function of IFN-γ in neuronal activity, renal sympathetic nerve activity (RSNA), and blood pressure (BP) regulation was determined through in-vivo and in-vitro experiments involving Ca 2+ imaging, immunofluorescence, platinum-iridium electrode recording, ELISA, the femoral artery cannulation test, and the tail-cuff method. RESULTS The BP, heart rate, RSNA, plasma norepinephrine, and the number of c-Fos-positive neurons in SIH rats increased compared with those in control rats. Pre and postsynaptic densities in the RVLM also increased in SIH rats. IFN-γ and CCL2 expression levels were significantly reduced in the RVLM of the SIH group, whose microglia also exhibited an impaired capacity for synapse engulfment. IFN-γ elevation increased CCL2 expression and microglial synaptic engulfment and decreased synaptic density in vivo and in vitro . However, CCL2 inhibition reversed these effects. Moreover, the reduction of neuronal excitability, RSNA, plasma norepinephrine, and BP by IFN-γ was abrogated through CCL2 expression. CONCLUSION IFN-γ deficiency in the RVLM impaired the microglial engulfment of synapses by inhibiting CCL2 expression and increasing synaptic density and neuronal excitability, thereby contributing to SIH progression. Targeting IFN-γ may be considered a potential strategy to combat SIH.
Collapse
Affiliation(s)
- Lei Tong
- College of Life Sciences, Shanghai University, Shanghai
| | - Gaojun Chen
- College of Life Sciences, Shanghai University, Shanghai
| | - Tianfeng Liu
- College of Life Sciences, Shanghai University, Shanghai
| | - Linping Wang
- College of Life Sciences, Shanghai University, Shanghai
| | - Haili Zhang
- College of Agriculture and Bioengineering, Heze University, Heze
| | - Fuxue Chen
- College of Life Sciences, Shanghai University, Shanghai
| | - Shuai Zhang
- International Cooperation Laboratory of Molecular Medicine, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou
| | - Dongshu Du
- College of Life Sciences, Shanghai University, Shanghai
- College of Agriculture and Bioengineering, Heze University, Heze
- Shaoxing Institute of Shanghai University, Shaoxing, China
| |
Collapse
|
26
|
Chen Z, Liang W, Liang J, Dou J, Guo F, Zhang D, Xu Z, Wang T. Probiotics: functional food ingredients with the potential to reduce hypertension. Front Cell Infect Microbiol 2023; 13:1220877. [PMID: 37465757 PMCID: PMC10351019 DOI: 10.3389/fcimb.2023.1220877] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 06/15/2023] [Indexed: 07/20/2023] Open
Abstract
Hypertension is an increasingly pressing public health concern across the globe. It can be triggered by a variety of factors such as age and diet, as well as the stress of modern life. The traditional treatment of hypertension includes calcium ion blockers, angiotensin II receptor inhibitors and β-receptor blockers, but these drugs have at least some side effects. Recent studies have revealed that intestinal flora plays a vital role in maintaining and promoting human health. This is due to the type and amount of probiotics present in the flora. Probiotics can reduce hypertension symptoms through four mechanisms: regulating vascular oxidative stress, producing short-chain fatty acids, restoring endothelial cell function, and reducing inflammation. It has been reported that certain functional foods, using probiotics as their raw material, can modify the composition of intestinal flora, thus regulating hypertension symptoms. Consequently, utilizing the probiotic function of probiotics in conjunction with the properties of functional foods to treat hypertension is a novel, side-effect-free treatment method. This study seeks to summarize the various factors that contribute to hypertension, the mechanism of probiotics in mitigating hypertension, and the fermented functional foods with probiotic strains, in order to provide a basis for the development of functional foods which utilize probiotics as their raw material and may have the potential to reduce hypertension.
Collapse
Affiliation(s)
- Zouquan Chen
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan, China
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan, China
| | - Wanjie Liang
- Research and Development Department(R&D), Shandong Ande Healthcare Apparatus Co., Ltd., Zibo, China
| | - Jie Liang
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan, China
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan, China
| | - Jiaxin Dou
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan, China
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan, China
| | - Fangyu Guo
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan, China
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan, China
| | - Daolei Zhang
- School of Bioengineering, Shandong Polytechnic, Jinan, China
- Henan Province Key Laboratory of Water Pollution Control and Rehabilitation Technology, Henan University of Urban Construction, Pingdingshan, China
| | - Zhenshang Xu
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan, China
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan, China
| | - Ting Wang
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan, China
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan, China
| |
Collapse
|
27
|
Chomanskis Ž, Jonkus V, Danielius T, Paulauskas T, Orvydaitė M, Melaika K, Rukšėnas O, Hendrixson V, Ročka S. Hypotensive Effect of Electric Stimulation of Caudal Ventrolateral Medulla in Freely Moving Rats. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1046. [PMID: 37374250 DOI: 10.3390/medicina59061046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023]
Abstract
Background and Objectives: An altered sympathetic function is established in primary arterial hypertension (PAH) development. Therefore, PAH could be targeted by applying an electric current to the medulla where reflex centers for blood pressure control reside. This study aims to evaluate the electric caudal ventrolateral medulla (CVLM) stimulation effect on blood pressure and animal survivability in a freely moving rat model. Materials and Methods: A total of 20 Wistar rats aged 12-16 weeks were randomly assigned to either: the experimental group (n = 10; electrode tip implanted in CVLM region) or the control group (n = 10; tip implanted 4 mm above the CVLM in the cerebellum). After a period of recovery (4 days), an experimental phase ensued, divided into an "OFF stimulation" period (5-7 days post-surgery) and an "ON stimulation" period (8-14 days post-surgery). Results: Three animals (15%, one in the control, two in the experimental group) dropped out due to postoperative complications. Arterial pressure in the experimental group rats during the "OFF stimulation" period decreased by 8.23 mm Hg (p = 0.001) and heart rate by 26.93 beats/min (p = 0.008). Conclusions: From a physiological perspective, CVLM could be an effective deep brain stimulation (DBS) target for drug-resistant hypertension: able to influence the baroreflex arc directly, having no known direct integrative or neuroendocrine function. Targeting the baroreflex regulatory center, but not its sensory or effector parts, could lead to a more predictable effect and stability of the control system. Although targeting neural centers in the medullary region is considered dangerous and prone to complications, it could open a new vista for deep brain stimulation therapy. A possible change in electrode design would be required to apply CVLM DBS in clinical trials in the future.
Collapse
Affiliation(s)
- Žilvinas Chomanskis
- Clinic of Neurology and Neurosurgery, Faculty of Medicine, Vilnius University, LT-01513 Vilnius, Lithuania
| | - Vytautas Jonkus
- Faculty of Physics, Vilnius University, LT-01513 Vilnius, Lithuania
| | - Tadas Danielius
- Institute of Applied Mathematics, Faculty of Mathematics and Informatics, Vilnius University, LT-01513 Vilnius, Lithuania
| | - Tomas Paulauskas
- Department of Neurobiology and Biophysics, Institute of Biosciences, Life Sciences Center, Vilnius University, LT-01513 Vilnius, Lithuania
| | - Monika Orvydaitė
- Faculty of Medicine, Vilnius University, LT-01513 Vilnius, Lithuania
| | | | - Osvaldas Rukšėnas
- Department of Neurobiology and Biophysics, Institute of Biosciences, Life Sciences Center, Vilnius University, LT-01513 Vilnius, Lithuania
| | - Vaiva Hendrixson
- Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, LT-01513 Vilnius, Lithuania
| | - Saulius Ročka
- Clinic of Neurology and Neurosurgery, Faculty of Medicine, Vilnius University, LT-01513 Vilnius, Lithuania
| |
Collapse
|
28
|
Liu T, Wang L, Chen G, Tong L, Ye X, Yang H, Liu H, Zhang H, Lu W, Zhang S, Du D. PDZD8-mediated endoplasmic reticulum-mitochondria associations regulate sympathetic drive and blood pressure through the intervention of neuronal mitochondrial homeostasis in stress-induced hypertension. Neurobiol Dis 2023:106173. [PMID: 37247681 DOI: 10.1016/j.nbd.2023.106173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 05/31/2023] Open
Abstract
Neuronal hyperexcitation in the rostral ventrolateral medulla (RVLM) drives heightened sympathetic nerve activity and contributes to the etiology of stress-induced hypertension (SIH). Maintenance of mitochondrial functions is central to neuronal homeostasis. PDZD8, an endoplasmic reticulum (ER) transmembrane protein, tethers ER to mitochondria. However, the mechanisms of PDZD8-mediated ER-mitochondria associations regulating neuronal mitochondrial functions and thereby mediating blood pressure (BP) in the RVLM of SIH were largely unknown. SIH rats were subjected to intermittent electric foot shocks plus noise for 2 h twice daily for 15 consecutive days. The underlying mechanisms of PDZD8 were investigated through in vitro experiments by using small interfering RNA and through in vivo experiments, such as intra-RVLM microinjection and Western blot analysis. The function of PDZD8 on BP regulation in the RVLM was determined in vivo via the intra-RVLM microinjection of adeno-associated virus (AAV)2-r-Pdzd8. We found that the c-Fos-positive RVLM tyrosine hydroxylase (TH) neurons, renal sympathetic nerve activity (RSNA), plasma norepinephrine (NE) level, BP, and heart rate (HR) were elevated in SIH rats. ER-mitochondria associations in RVLM neurons were significantly reduced in SIH rats. PDZD8 was mainly expressed in RVLM neurons, and mRNA and protein levels were markedly decreased in SIH rats. In N2a cells, PDZD8 knockdown disrupted ER-mitochondria associations and mitochondrial structure, decreased mitochondrial membrane potential (MMP) and respiratory metabolism, enhanced ROS levels, and reduced catalase (CAT) activity. These effects suggested that PDZD8 dysregulation induced mitochondrial malfunction. By contrast, PDZD8 upregulation in the RVLM of SIH rats could rescue neuronal mitochondrial function, thereby suppressing c-Fos expression in TH neurons and decreasing RSNA, plasma NE, BP, and HR. Our results indicated that the dysregulation of PDZD8-mediated ER-mitochondria associations led to the loss of the activity homeostasis of RVLM neurons by disrupting mitochondrial functions, thereby participating in the regulation of SIH pathology.
Collapse
Affiliation(s)
- Tianfeng Liu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China; College of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Linping Wang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China; College of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Gaojun Chen
- College of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Lei Tong
- College of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Xuanxuan Ye
- College of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Hui Yang
- College of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Haisheng Liu
- College of Agriculture and Bioengineering, Heze University, Heze 274000, China
| | - Haili Zhang
- College of Agriculture and Bioengineering, Heze University, Heze 274000, China
| | - Wen Lu
- College of Agriculture and Bioengineering, Heze University, Heze 274000, China
| | - Shuai Zhang
- International Cooperation Laboratory of Molecular Medicine, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Dongshu Du
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China; College of Life Sciences, Shanghai University, Shanghai 200444, China; Shaoxing Institute of Shanghai University, Shaoxing, Zhejiang 312000, China; College of Agriculture and Bioengineering, Heze University, Heze 274000, China.
| |
Collapse
|
29
|
Gomes PM, Batista JS, Sá RWM, Antunes VR. Short exposure to high salt in drinking solution leads to a cardiovascular phenotype of hypertension without changes in the blood volume of rats. Exp Physiol 2023; 108:361-370. [PMID: 36715005 PMCID: PMC10103861 DOI: 10.1113/ep090912] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 01/06/2023] [Indexed: 01/31/2023]
Abstract
NEW FINDINGS What is the central question of this study? Is the cardiovascular phenotype of high blood pressure observed in rats salt loaded with 2% NaCl in drinking solution a blood volume-dependent hypertension? What is the main finding and its importance? Animals exposed to 2% NaCl drinking solution develop hypertension, with dominance of sympathetic outflow and high [Na+ ] in the cerebrospinal fluid, but without changes in the blood volume. The phenotype of salt-dependent hypertension might be related to accumulation of [Na+ ] in the cerebrospinal fluid, which makes it an interesting animal model in which to study the neuronal pathways involved in control of the circulation in osmotic challenge conditions. ABSTRACT Evidence suggests that hypertension induced by high salt intake is correlated with an autonomic imbalance that favours sympathetic hyperactivity and an increase in vascular resistance, indicating a neurogenic component to this pathology. Although there are several animal models in which to study salt-induced hypertension with prolonged exposure to a high-sodium diet, here we sought to investigate whether the increase in arterial blood pressure of rats subjected to a short exposure to high salt, with 2% NaCl drinking solution instead of water, relies on changes in the circulating blood volume. Male Wistar rats were divided randomly into three groups: euhydrated (EU, n = 10), salt loaded (SL, n = 13) and water deprived (WD, n = 6). The SL rats exhibited a significant increase in mean arterial blood pressure, with a large low-frequency component of systolic arterial blood pressure variability, when compared with the EU group. Circulating blood volume did not differ between SL and EU rats, but it was lower in WD rats. Compared with EU rats, the [Na+ ] in cerebrospinal fluid was higher in SL rats and similar in magnitude to the WD rats. Plasma [Na+ ] did not differ between SL and EU rats, but it was higher in WD rats. Collectively, our data suggest that the hypertension induced by a short exposure to high salt intake closely resembles a neurogenic mechanism, but not a blood volume-dependent mechanism, with cumulative [Na+ ] in the cerebrospinal fluid that could be associated with changes in the neurochemistry of autonomic nuclei, which are highly susceptible to osmotic stress related to high salt consumption.
Collapse
Affiliation(s)
- Paula Magalhães Gomes
- Department of Physiology and BiophysicsInstitute of Biomedical SciencesUniversity of Sao PauloSao PauloSPBrazil
| | - Julia Santos Batista
- Department of Physiology and BiophysicsInstitute of Biomedical SciencesUniversity of Sao PauloSao PauloSPBrazil
| | - Renato Willian Martins Sá
- Department of Physiology and BiophysicsInstitute of Biomedical SciencesUniversity of Sao PauloSao PauloSPBrazil
| | - Vagner Roberto Antunes
- Department of Physiology and BiophysicsInstitute of Biomedical SciencesUniversity of Sao PauloSao PauloSPBrazil
| |
Collapse
|
30
|
Central control of cardiac activity as assessed by intra-cerebral recordings and stimulations. Neurophysiol Clin 2023; 53:102849. [PMID: 36867969 DOI: 10.1016/j.neucli.2023.102849] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 03/05/2023] Open
Abstract
Some of the most important integrative control centers for the autonomic nervous system are located in the brainstem and the hypothalamus. However, growing recent neuroimaging evidence support that a set of cortical regions, named the central autonomic network (CAN), is involved in autonomic control and seems to play a major role in continuous autonomic cardiac adjustments to high-level emotional, cognitive or sensorimotor cortical activities. Intracranial explorations during stereo-electroencephalography (SEEG) offer a unique opportunity to address the question of the brain regions involved in heart-brain interaction, by studying: (i) direct cardiac effects produced by the electrical stimulation of specific brain areas; (ii) epileptic seizures inducing cardiac modifications; (iii) cortical regions involved in cardiac interoception and source of cardiac evoked potentials. In this review, we detail the available data assessing cardiac central autonomic regulation using SEEG, address the strengths and also the limitations of this technique in this context, and discuss perspectives. The main cortical regions that emerge from SEEG studies as being involved in cardiac autonomic control are the insula and regions belonging to the limbic system: the amygdala, the hippocampus, and the anterior and mid-cingulate. Although many questions remain, SEEG studies have already demonstrated afferent and efferent interactions between the CAN and the heart. Future studies in SEEG should integrate these afferent and efferent dimensions as well as their interaction with other cortical networks to better understand the functional heart-brain interaction.
Collapse
|
31
|
Renal sympathetic activity: A key modulator of pressure natriuresis in hypertension. Biochem Pharmacol 2023; 208:115386. [PMID: 36535529 DOI: 10.1016/j.bcp.2022.115386] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Hypertension is a complex disorder ensuing necessarily from alterations in the pressure-natriuresis relationship, the main determinant of long-term control of blood pressure. This mechanism sets natriuresis to the level of blood pressure, so that increasing pressure translates into higher osmotically driven diuresis to reduce volemia and control blood pressure. External factors affecting the renal handling of sodium regulate the pressure-natriuresis relationship so that more or less natriuresis is attained for each level of blood pressure. Hypertension can thus only develop following primary alterations in the pressure to natriuresis balance, or by abnormal activity of the regulation network. On the other hand, increased sympathetic tone is a very frequent finding in most forms of hypertension, long regarded as a key element in the pathophysiological scenario. In this article, we critically analyze the interplay of the renal component of the sympathetic nervous system and the pressure-natriuresis mechanism in the development of hypertension. A special focus is placed on discussing recent findings supporting a role of baroreceptors as a component, along with the afference of reno-renal reflex, of the input to the nucleus tractus solitarius, the central structure governing the long-term regulation of renal sympathetic efferent tone.
Collapse
|
32
|
Lamptey RNL, Sun C, Layek B, Singh J. Neurogenic Hypertension, the Blood-Brain Barrier, and the Potential Role of Targeted Nanotherapeutics. Int J Mol Sci 2023; 24:2213. [PMID: 36768536 PMCID: PMC9916775 DOI: 10.3390/ijms24032213] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023] Open
Abstract
Hypertension is a major health concern globally. Elevated blood pressure, initiated and maintained by the brain, is defined as neurogenic hypertension (NH), which accounts for nearly half of all hypertension cases. A significant increase in angiotensin II-mediated sympathetic nervous system activity within the brain is known to be the key driving force behind NH. Blood pressure control in NH has been demonstrated through intracerebrovascular injection of agents that reduce the sympathetic influence on cardiac functions. However, traditional antihypertensive agents lack effective brain permeation, making NH management extremely challenging. Therefore, developing strategies that allow brain-targeted delivery of antihypertensives at the therapeutic level is crucial. Targeting nanotherapeutics have become popular in delivering therapeutics to hard-to-reach regions of the body, including the brain. Despite the frequent use of nanotherapeutics in other pathological conditions such as cancer, their use in hypertension has received very little attention. This review discusses the underlying pathophysiology and current management strategies for NH, as well as the potential role of targeted therapeutics in improving current treatment strategies.
Collapse
Affiliation(s)
| | | | - Buddhadev Layek
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND 58105, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND 58105, USA
| |
Collapse
|
33
|
A C, R C, N B, G DI. Compression therapy, autonomic nervous system, and heart rate variability: A narrative review and our preliminary personal experience. Phlebology 2022; 37:739-753. [DOI: 10.1177/02683555221135321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aims To highlight the relationship among compression therapy (CT), the autonomic nervous system (ANS) (parasympathetic and sympathetic system), and the heart rate variability (HRV) analysis. Background Beyond the typical analgesic and anti-inflammatory effects of CT in patients affected by venous and/or lymphatic diseases, some literature about CT influence on wellbeing has been published as well. More specifically, CT influence on the ANS has been elucidated mostly through HRV application, providing useful quali-quantitative data for scientific and clinical purposes. Material and Methods A literature search was performed through several web-based search engines to investigate the available evidence concerning the possible influence of CT on the ANS and on psychoneuroendocrineimmunology. Moreover, we examined literature data regarding HRV use in the assessment of CT. Lastly, a preliminary cross-over study was performed on 10 patients affected by phlebolymphedema of the lower limbs, undergoing CT with 18–21 mmHg stockings for 10 h and investigated by means of HRV. Results A CT-based increase of the anti-inflammatory activity of the parasympathetic (vagal) system has been elucidated in most scientific literature. Similarly, CT application has generally resulted in an improvement of HRV, which indicates a beneficial influence on the ANS. In our preliminary experience with compression stockings and HRV, two parasympathetic-based parameters improved by 22.8% and 68.0% after 10 h, whereas they decreased in the same subjects without stockings by 2.7% and 8.2%, during normal breathing. The remaining HRV parameters did not show relevant variations, especially during diaphragmatic breathing. Conclusions From literature data and based on our very preliminary experience, it is possible to deduce that CT exerts different effects on the psychobiological parameters of the individual, overall improving HRV and parasympathetic activity. Incorporating both HRV/ANS assessment in phlebolymphology and the beneficial neural action of CT in health care may represent viable options in the future biomedical science.
Collapse
Affiliation(s)
- Cavezzi A
- Eurocenter Venalinfa, San Benedetto Del Tronto (AP), Italy
| | - Colucci R
- Eurocenter Venalinfa, San Benedetto Del Tronto (AP), Italy
| | - Barsotti N
- CMO-Centro di Medicina Osteopatica, Firenze, Italy
| | - Di Ionna G
- Strategic Nutrition Center, Bologna, Italy
| |
Collapse
|
34
|
Jami O, Tijani Y, Et-Tahir A. Device-Based Therapy for Resistant Hypertension: An Up-to-Date Review. High Blood Press Cardiovasc Prev 2022; 29:537-546. [PMID: 36178479 PMCID: PMC9523625 DOI: 10.1007/s40292-022-00539-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 08/23/2022] [Indexed: 11/29/2022] Open
Abstract
Hypertension is the major risk factor for cardiovascular morbidity and mortality. Matter of fact, untreated hypertension can worsen the overall health, whereas pharmacotherapy can play an important role in lowering the risk of high blood pressure in hypertensive patients. However, persistent uncontrolled hypertension remains an unsolved condition characterized by non-adherence to medication and increased sympathetic activity. This paper will review the non-pharmacological treatments for resistant hypertension (RH) that have emerged in recent years. In addition, the technologies developed in device-based RH therapy, as well as the clinical trials that support their use, will be discussed. Indeed, the novel device-based approaches that target RH present a promising therapy which has been supported by several studies and clinical trials, whereas drug non-adherence and high sympathetic activity are known to be the main causes of RH. Nevertheless, some additional aspects of these RH systems need to be tested in the near future, with a particular focus on the device's design and availability of randomized controlled trials.
Collapse
Affiliation(s)
- Oussama Jami
- High School of Technology in Salé, Materials, Energy and Acoustics Team Rabat, Mohammed V University in Rabat, Rabat, Morocco.
- Biomedical Engineering Department, Mohammed VI University of Health Sciences, Casablanca, Morocco.
| | - Youssef Tijani
- Faculty of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Aziz Et-Tahir
- High School of Technology in Salé, Materials, Energy and Acoustics Team Rabat, Mohammed V University in Rabat, Rabat, Morocco
| |
Collapse
|
35
|
Yang ZJ, Wang TT, Wang BY, Gao H, He CW, Shang HW, Lu X, Wang Y, Xu JD. Deeper insight into the role of IL-17 in the relationship beween hypertension and intestinal physiology. J Inflamm (Lond) 2022; 19:14. [PMID: 36195874 PMCID: PMC9530412 DOI: 10.1186/s12950-022-00311-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 09/21/2022] [Indexed: 11/10/2022] Open
Abstract
With the incidence of hypertension increasing worldwide, more and more the mechanisms of hypertension from the perspective of immunity have found. Intestinal microbiota as well as its metabolites relationship with hypertension has attracted great attention from both clinicians and investigators. However, the associations of hypertension with lesions of a large number of immune factors including IL-17, MCP-1, IL-6, TGF-β, IL-10 and others have not been fully characterized. In this review, after introducing the immune factors as the most potent anti/pro-hypertension agents known, we provide detailed descriptions of the IL-17 involved in the pathology of hypertension, pointing out the underlying mechanisms and suggesting the clinical indications.
Collapse
Affiliation(s)
- Ze-Jun Yang
- grid.24696.3f0000 0004 0369 153XClinical Medicine of “5+3”program, School of Basic Medical Science, Capital Medical University, Beijing, China ,grid.24696.3f0000 0004 0369 153XDepartment of Cardiology, Beijing An Zhen Hospital, Capital Medical University, Beijing, China
| | - Tian-Tian Wang
- grid.24696.3f0000 0004 0369 153XDepartment of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Bo-Ya Wang
- grid.411634.50000 0004 0632 4559Eight Program of Clinical Medicine, Peking University People’s Hospital, Beijing, China
| | - Han Gao
- grid.24696.3f0000 0004 0369 153XDepartment of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Cheng-Wei He
- grid.24696.3f0000 0004 0369 153XDepartment of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Hong-Wei Shang
- grid.24696.3f0000 0004 0369 153XMorphological Experiment Center, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xin Lu
- grid.24696.3f0000 0004 0369 153XMorphological Experiment Center, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ying Wang
- grid.414373.60000 0004 1758 1243Department of Dermatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jing-Dong Xu
- grid.24696.3f0000 0004 0369 153XDepartment of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
36
|
Exercise Training Attenuates Hypertension via Suppressing ROS/MAPK/NF-κB/AT-1R Pathway in the Hypothalamic Paraventricular Nucleus. Nutrients 2022; 14:nu14193968. [PMID: 36235619 PMCID: PMC9573547 DOI: 10.3390/nu14193968] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Aerobic exercise training (ExT) is beneficial for hypertension, however, its central mechanisms in improving hypertension remain unclear. Since the importance of the up-regulation of angiotensin II type 1 receptor (AT-1R) in the paraventricular nucleus (PVN) of the hypothalamic in sympathoexcitation and hypertension has been shown, we testified the hypothesis that aerobic ExT decreases blood pressure in hypertensive rats by down-regulating the AT-1R through reactive oxygen species (ROS)/mitogen-activated protein kinase (MAPK)/nuclear factors κB (NF-κB) pathway within the PVN. Methods: Forty-eight male Sprague-Dawley (SD) rats were assigned to the following groups: sham operation (SHAM) + kept sedentary (Sed), SHAM + exercise training (ExT), two kidney-one clamp (2K1C) + Sed, and 2K1C + ExT groups. Results: The 2K1C + Sed hypertensive rats showed higher systolic blood pressure (SBP), upregulated ROS, phosphorylated (p-) p44/42 MAPK, p-p38 MAPK, NF-κB p65 activity, and AT-1R expression in the PVN, and increased circulating norepinephrine (NE) than those of SHAM rats. After eight weeks of aerobic ExT, the 2K1C + ExT hypertensive rats showed attenuated NE and SBP levels, suppressed NF-κB p65 activity, and reduced expression of ROS, p-p44/42 MAPK, p-p38 MAPK, and AT-1R in the PVN, relatively to the 2K1C + Sed group. Conclusions: These data are suggestive of beneficial effects of aerobic ExT in decreasing SBP in hypertensive rats, via down-regulating the ROS/MAPK/NF-κB pathway that targets AT-1R in the PVN, and eventually ameliorating 2K1C-induced hypertension.
Collapse
|
37
|
Li J, Zhou L, Gong H. New insights and advances of sodium-glucose cotransporter 2 inhibitors in heart failure. Front Cardiovasc Med 2022; 9:903902. [PMID: 36186974 PMCID: PMC9520058 DOI: 10.3389/fcvm.2022.903902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2is) are newly emerging insulin-independent anti-hyperglycemic agents that work independently of β-cells. Quite a few large-scale clinical trials have proven the cardiovascular protective function of SGLT2is in both diabetic and non-diabetic patients. By searching all relevant terms related to our topics over the previous 3 years, including all the names of agents and their brands in PubMed, here we review the mechanisms underlying the improvement of heart failure. We also discuss the interaction of various mechanisms proposed by diverse works of literature, including corresponding and opposing viewpoints to support each subtopic. The regulation of diuresis, sodium excretion, weight loss, better blood pressure control, stimulation of hematocrit and erythropoietin, metabolism remodeling, protection from structural dysregulation, and other potential mechanisms of SGLT2i contributing to heart failure improvement have all been discussed in this manuscript. Although some remain debatable or even contradictory, those newly emerging agents hold great promise for the future in cardiology-related therapies, and more research needs to be conducted to confirm their functionality, particularly in metabolism, Na+-H+ exchange protein, and myeloid angiogenic cells.
Collapse
Affiliation(s)
- Juexing Li
- Department of Cardiology, Jinshan Hospital of Fudan University, Shanghai, China
- Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Zhou
- Department of Cardiology, Jinshan Hospital of Fudan University, Shanghai, China
- Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Gong
- Department of Cardiology, Jinshan Hospital of Fudan University, Shanghai, China
- Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Hui Gong
| |
Collapse
|
38
|
Chao YM, Rauchová H, Chan JYH. Disparate Roles of Oxidative Stress in Rostral Ventrolateral Medulla in Age-Dependent Susceptibility to Hypertension Induced by Systemic l-NAME Treatment in Rats. Biomedicines 2022; 10:biomedicines10092232. [PMID: 36140333 PMCID: PMC9496567 DOI: 10.3390/biomedicines10092232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 12/12/2022] Open
Abstract
This study aims to investigate whether tissue oxidative stress in the rostral ventrolateral medulla (RVLM), where sympathetic premotor neurons reside, plays an active role in age-dependent susceptibility to hypertension in response to nitric oxide (NO) deficiency induced by systemic l-NAME treatment, and to decipher the underlying molecular mechanisms. Systolic blood pressure (SBP) and heart rate (HR) in conscious rats were recorded, along with measurements of plasma and RVLM level of NO and reactive oxygen species (ROS), and expression of mRNA and protein involved in ROS production and clearance, in both young and adult rats subjected to intraperitoneal (i.p.) infusion of l-NAME. Pharmacological treatments were administered by oral gavage or intracisternal infusion. Gene silencing of target mRNA was made by bilateral microinjection into RVLM of lentivirus that encodes a short hairpin RNA (shRNA) to knock down gene expression of NADPH oxidase activator 1 (Noxa1). We found that i.p. infusion of l-NAME resulted in increases in SBP, sympathetic neurogenic vasomotor activity, and plasma norepinephrine levels in an age-dependent manner. Systemic l-NAME also evoked oxidative stress in RVLM of adult, but not young rats, accompanied by augmented enzyme activity of NADPH oxidase and reduced mitochondrial electron transport enzyme activities. Treatment with L-arginine via oral gavage or infusion into the cistern magna (i.c.), but not i.c. tempol or mitoQ10, significantly offset the l-NAME-induced hypertension in young rats. On the other hand, all treatments appreciably reduced l-NAME-induced hypertension in adult rats. The mRNA microarray analysis revealed that four genes involved in ROS production and clearance were differentially expressed in RVLM in an age-related manner. Of them, Noxa1, and GPx2 were upregulated and Duox2 and Ucp3 were downregulated. Systemic l-NAME treatment caused greater upregulation of Noxa1, but not Ucp3, mRNA expression in RVLM of adult rats. Gene silencing of Noxa1 in RVLM effectively alleviated oxidative stress and protected adult rats against l-NAME-induced hypertension. These data together suggest that hypertension induced by systemic l-NAME treatment in young rats is mediated primarily by NO deficiency that occurs both in vascular smooth muscle cells and RVLM. On the other hand, enhanced augmentation of oxidative stress in RVLM may contribute to the heightened susceptibility of adult rats to hypertension induced by systemic l-NAME treatment.
Collapse
Affiliation(s)
- Yung-Mei Chao
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Hana Rauchová
- Institute of Physiology, Czech Academy of Sciences, 14200 Prague, Czech Republic
| | - Julie Y. H. Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Correspondence: ; Tel.: +886-77338415
| |
Collapse
|
39
|
Li X, Yin Z, Ling F, Zheng Q, Li X, Qi W, Liang F. The application of acupuncture in cardiopathy: A bibliometric analysis based on Web of Science across ten recent years. Front Cardiovasc Med 2022; 9:920491. [PMID: 36148057 PMCID: PMC9485815 DOI: 10.3389/fcvm.2022.920491] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Background With high morbidity and mortality, cardiopathy is a major component of cardiovascular disease, causing a huge burden of disease to public health worldwide. In recent years, research on acupuncture treatment of cardiopathies has been increasing. However, no bibliometric analysis has been conducted to systematically describe the research progress and hotspots in this field. Therefore, this study aimed to conduct a bibliometric analysis of the relevant literature to explore the current status and future development of acupuncture for cardiopathies. Methods The Web of Science (WoS) Core Collection Database was searched for literature related to acupuncture therapies for cardiopathies from 2011 to 2021. Using CiteSpace 5.8 R3, cooperation network diagrams of authors, institutions, countries and journals, keyword co-occurrences, and clustering were performed and analyzed. Results A total of 321 studies were included. Overall, the number of annual publications increased yearly. These publications came from 31 countries or regions, of which China and the United States made the greatest contributions. In total, 333 authors from 258 institutions participated in this field, and Beijing University of Chinese Medicine and Professor Fanrong Liang were the most published institution and author, respectively. Evidence-based Complementary and Alternative Medicine published the largest number of articles, and CIRCULATION was the most commonly cited journal. Based on co-occurrences and cluster analysis of 257 keywords, three research frontiers and hotspots were identified: acupuncture for blood pressure regulation, acupuncture for coronary heart disease, and acupuncture for regulation of heart rate. In these three research frontiers, the rostral ventrolateral medulla (RVLM) and autonomic nervous system (ANS) are the most popular mechanisms. Conclusion A stable development trend has formed in this field. Further research should focus on the role of acupuncture therapies in the treatment of hypertension or hypertensive heart disease, coronary heart disease, and arrhythmia based on the mechanisms related to the RVLM and ANS.
Collapse
Affiliation(s)
- Xiao Li
- School of Acu-Mox and Tuina/The Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zihan Yin
- School of Acu-Mox and Tuina/The Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Acupuncture and Tuina School/The Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine/Clinical Research Center for Acupuncture and Moxibustion in Sichuan Province, Chengdu, Sichuan, China
| | - Fayang Ling
- School of Acu-Mox and Tuina/The Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qianhua Zheng
- School of Acu-Mox and Tuina/The Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiang Li
- School of Acu-Mox and Tuina/The Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- *Correspondence: Xiang Li,
| | - Wenchuan Qi
- School of Acu-Mox and Tuina/The Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Wenchuan Qi,
| | - Fanrong Liang
- School of Acu-Mox and Tuina/The Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Acupuncture and Tuina School/The Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine/Clinical Research Center for Acupuncture and Moxibustion in Sichuan Province, Chengdu, Sichuan, China
- Fanrong Liang,
| |
Collapse
|
40
|
Chemogenetic inhibition of Phox2-expressing neurons in the commissural NTS decreases blood pressure in anesthetized spontaneously hypertensive rats. Neurosci Lett 2022; 787:136817. [PMID: 35905886 DOI: 10.1016/j.neulet.2022.136817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 07/17/2022] [Accepted: 07/22/2022] [Indexed: 11/21/2022]
Abstract
Interruption of the activity of neurons in the commissural portion of the nucleus of the solitary tract (cNTS) decreases blood pressure (BP) in experimental models of hypertension, such as the spontaneously hypertensive (SH) rat. To examine whether PHOX2B expressing cNTS neurons are involved in maintaining the elevated BP, we used replication-deficient viruses with a modified Phox2 binding site promoter to express the inhibitory chemogenetic allatostatin receptor or green fluorescent protein in the cNTS. Following administration of allatostatin, we observed a depressor and bradycardic response in anesthetized SH rats that expressed the allatostatin receptor. Injection of allatostatin did not affect BP or heart rate (HR) in control SH rats expressing green fluorescent protein in the cNTS. Immunohistochemistry showed that the majority of transduced cNTS neurons were PHOX2B-immunoreactive and some also expressed tyrosine hydroxylase. We conclude that in anesthetized SH rat, the Phox2B expressing cNTS neurons maintain elevated BP.
Collapse
|
41
|
Irisin, An Exercise-induced Bioactive Peptide Beneficial for Health Promotion During Aging Process. Ageing Res Rev 2022; 80:101680. [DOI: 10.1016/j.arr.2022.101680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 12/11/2022]
|
42
|
Liu G, Cheng J, Zhang T, Shao Y, Chen X, Han L, Zhou R, Wu B. Inhibition of Microbiota-dependent Trimethylamine N-Oxide Production Ameliorates High Salt Diet-Induced Sympathetic Excitation and Hypertension in Rats by Attenuating Central Neuroinflammation and Oxidative Stress. Front Pharmacol 2022; 13:856914. [PMID: 35359866 PMCID: PMC8961329 DOI: 10.3389/fphar.2022.856914] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/14/2022] [Indexed: 12/18/2022] Open
Abstract
Excessive dietary salt intake induces neuroinflammation and oxidative stress in the brain, which lead to sympathetic excitation, contributing to hypertension. However, the underlying mechanisms remain elusive. Accumulating evidence reveals that trimethylamine-N-oxide (TMAO), a gut microbiota-derived metabolite, is implicated in the pathogenesis of multiple cardiovascular diseases. The present study sought to determine whether central TMAO is elevated and associated with neuroinflammation and oxidative stress in the brain after long-term high salt (HS) diet intake and, if so, whether inhibition of TMAO generation ameliorates HS-induced sympathetic excitation and hypertension. Sprague-Dawley rats were fed either a HS diet or a normal salt (NS) diet and simultaneously treated with vehicle (VEH) or 1.0% 3,3-Dimethyl-1-butanol (DMB, an inhibitor of trimethylamine formation) for 8 weeks. HS + VEH rats, compared with NS + VEH rats, had elevated TMAO in plasma and cerebrospinal fluid (CSF), increased blood pressure (BP), and increased sympathetic drive as indicated by the BP response to ganglionic blockade and plasma norepinephrine levels. HS-induced these changes were attenuated by DMB, which significantly reduced TMAO in plasma and CSF. Neuroinflammation as assessed by proinflammatory cytokine expression and NF-κB activity and microglial activity, and oxidative stress as measured by NAD(P)H oxidase subunit expression and NAD(P)H activity and reactive oxygen species (ROS) production in the hypothalamic paraventricular nucleus (PVN) were increased in HS + VEH rats but were decreased by DMB. DMB had no effects on above measured parameters in NS rats. The results suggest that long-term HS diet intake causes elevation in TMAO in the circulation and brain, which is associated with increased neuroinflammation and oxidative stress in the PVN, an important cardiovascular regulatory center. Inhibition of TMAO generation ameliorates HS-induced sympathetic excitation and hypertension by reducing neuroinflammation and oxidative stress in the PVN.
Collapse
Affiliation(s)
- Gang Liu
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Jiayin Cheng
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Tianhao Zhang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Yingxin Shao
- Department of General Practice, The First Hospital of China Medical University, Shenyang, China
| | - Xiangxu Chen
- Department of General Practice, The First Hospital of China Medical University, Shenyang, China
| | - Lihong Han
- Department of General Practice, The First Hospital of China Medical University, Shenyang, China
| | - Ru Zhou
- Department of General Practice, The First Hospital of China Medical University, Shenyang, China
| | - Bin Wu
- Department of General Practice, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
43
|
Fossey MPM, Balthazaar SJT, Squair JW, Williams AM, Poormasjedi-Meibod MS, Nightingale TE, Erskine E, Hayes B, Ahmadian M, Jackson GS, Hunter DV, Currie KD, Tsang TSM, Walter M, Little JP, Ramer MS, Krassioukov AV, West CR. Spinal cord injury impairs cardiac function due to impaired bulbospinal sympathetic control. Nat Commun 2022; 13:1382. [PMID: 35296681 PMCID: PMC8927412 DOI: 10.1038/s41467-022-29066-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 02/20/2022] [Indexed: 02/08/2023] Open
Abstract
Spinal cord injury chronically alters cardiac structure and function and is associated with increased odds for cardiovascular disease. Here, we investigate the cardiac consequences of spinal cord injury on the acute-to-chronic continuum, and the contribution of altered bulbospinal sympathetic control to the decline in cardiac function following spinal cord injury. By combining experimental rat models of spinal cord injury with prospective clinical studies, we demonstrate that spinal cord injury causes a rapid and sustained reduction in left ventricular contractile function that precedes structural changes. In rodents, we experimentally demonstrate that this decline in left ventricular contractile function following spinal cord injury is underpinned by interrupted bulbospinal sympathetic control. In humans, we find that activation of the sympathetic circuitry below the level of spinal cord injury causes an immediate increase in systolic function. Our findings highlight the importance for early interventions to mitigate the cardiac functional decline following spinal cord injury. By combining experimental models with prospective clinical studies, the authors show that spinal cord injury causes a rapid reduction in cardiac function that precedes structural changes, and that the loss of descending sympathetic control is the major cause of reduced cardiac function following spinal cord injury.
Collapse
Affiliation(s)
- Mary P M Fossey
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada.,Experimental Medicine, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Shane J T Balthazaar
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada.,Experimental Medicine, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jordan W Squair
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
| | - Alexandra M Williams
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada.,Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Tom E Nightingale
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada.,School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK.,Centre for Trauma Sciences Research, University of Birmingham, Edgabaston, Birmingham, UK
| | - Erin Erskine
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada.,Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Brian Hayes
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
| | - Mehdi Ahmadian
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada.,School of Kinesiology, Faculty of Education, University of British Columbia, Vancouver, BC, Canada
| | - Garett S Jackson
- Faculty of Health and Social Development, University of British Columbia, Kelowna, BC, Canada
| | - Diana V Hunter
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
| | - Katharine D Currie
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
| | - Teresa S M Tsang
- Division of Cardiology, University of British Columbia, Vancouver General and University of British Columbia Hospital Echocardiography Department, Vancouver, BC, Canada
| | - Matthias Walter
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada.,Department of Urology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Jonathan P Little
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| | - Matt S Ramer
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada.,Department of Zoology, Faculty of Science, University of British Columbia, Vancouver, BC, Canada
| | - Andrei V Krassioukov
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada. .,Experimental Medicine, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada. .,Division of Physical Medicine and Rehabilitation, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada. .,GF Strong Rehabilitation Centre, Vancouver Coastal Health, Vancouver, BC, Canada.
| | - Christopher R West
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada. .,Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
44
|
Abstract
Hypertension is a worldwide problem with major impacts on health including morbidity and mortality, as well as consumption of health care resources. Nearly 50% of American adults have high blood pressure, and this rate is rising. Even with multiple antihypertensive drugs and aggressive lifestyle modifications, blood pressure is inadequately controlled in about 1 of 5 hypertensive individuals. This review highlights a hypothesis for hypertension that suggests alternative mechanisms for blood pressure elevation and maintenance. A better understanding of these mechanisms could open avenues for more successful treatments. The hypothesis accounts for recent understandings of the involvement of gut physiology, gut microbiota, and neuroinflammation in hypertension. It includes bidirectional communication between gut microbiota and gut epithelium in the gut-brain axis that is involved in regulation of autonomic nervous system activity and blood pressure control. Dysfunction of this gut-brain axis, including dysbiosis of gut microbiota, gut epithelial dysfunction, and deranged input to the brain, contributes to hypertension via inflammatory mediators, metabolites, bacteria in the circulation, afferent information alterations, etc resulting in neuroinflammation and unbalanced autonomic nervous system activity that elevates blood pressure. This in turn negatively affects gut function and its microbiota exacerbating the problem. We focus this review on the gut-brain axis hypothesis for hypertension and possible contribution to racial disparities in hypertension. A novel idea, that immunoglobulin A-coated bacteria originating in the gut with access to the brain could be involved in hypertension, is raised. Finally, minocycline, with its anti-inflammatory and antimicrobial properties, is evaluated as a potential antihypertensive drug acting on this axis.
Collapse
Affiliation(s)
- Elaine M Richards
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Jing Li
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Bruce R Stevens
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Carl J Pepine
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Mohan K Raizada
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
45
|
Involvement of Neuropeptide Y within Paraventricular Nucleus in Electroacupuncture Inhibiting Sympathetic Activities in Hypertensive Rats. Int J Hypertens 2022; 2022:9990854. [PMID: 35087687 PMCID: PMC8789434 DOI: 10.1155/2022/9990854] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/05/2022] [Indexed: 12/14/2022] Open
Abstract
Although electroacupuncture (EA) has been used to decrease the blood pressure (BP) clinically, the underlying mechanisms are not clearly clarified. This study aimed to assess the hypothesis that EA treatment exerts a hypotensive action via suppressing sympathetic activities and modulating neuropeptide Y (NPY) function within the paraventricular nucleus (PVN) of hypertensive rats. Male Sprague-Dawley rats were selected for the experiment, and the hypertensive models were established by the two-kidney, one-clip (2K1C) method. Then, the rats were randomly assigned to the sham group, 2K1C group, 2K1C plus EA group, and 2K1C plus sham EA group. EA treatment at the acupoints ST36 and ST40 overlying the peroneal nerves was given once a day for 30 days. The radiotelemetry system was applied to collect the arterial BP recordings. Power spectral analyses of BP variability, BP responses to ganglionic blockade, and plasma levels of norepinephrine and epinephrine were performed to assess the changes in sympathetic nerve activity. Real-time PCR and Western blots were carried out to examine the expression of NPY system in the PVN. The responses of PVN microinjection with NPY Y1R antagonist BIBO3304 were detected to check the endogenous NPY tone. The results showed that the enhanced arterial BP and sympathetic activities were effectively reduced by 30 days of EA treatment, and baroreflex sensitivity was improved in 2K1C hypertensive rats. The level of NPY mRNA and protein expression in the PVN was markedly upregulated by EA treatment in 2K1C rats. In addition, the pressor responses of PVN microinjection with NPY Y1R antagonist BIBO3304 in 2K1C models were remarkably augmented by the EA stimulation. Our results indicate that the increased NPY expression and function in the PVN induced by EA treatment contribute to antihypertensive and sympathetic suppression on hypertensive rats. The findings may elucidate the underlying mechanisms of the acupuncture to be a potential therapeutic strategy against hypertension.
Collapse
|
46
|
Stocker SD, Wenner MM, Farquhar WB, Browning KN. Activation of the Organum Vasculosum of the Lamina Terminalis Produces a Sympathetically Mediated Hypertension. Hypertension 2022; 79:139-149. [PMID: 34809435 PMCID: PMC8665096 DOI: 10.1161/hypertensionaha.121.18117] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Neurons in the organum vasculosum of the lamina terminalis (OVLT) sense extracellular NaCl and angiotensin II concentrations to regulate body fluid homeostasis and arterial blood pressure. Lesion of the anteroventral third ventricular region or OVLT attenuates multiple forms of neurogenic hypertension. However, the extent by which OVLT neurons directly regulate sympathetic nerve activity to produce hypertension is not known. Therefore, the present study tested this hypothesis by using a multi-faceted approach including optogenetics, single-unit and multifiber nerve recordings, and chemogenetics. First, optogenetic activation of OVLT neurons in conscious Sprague-Dawley rats (250-400 g) produced frequency-dependent increases in arterial blood pressure and heart rate. These responses were not altered by the vasopressin receptor antagonist (β-mercapto-β,β-cyclopentamethylenepropionyl1,O-me-Tyr2,Arg8)-vasopressin but eliminated by the ganglionic blocker chlorisondamine. Second, optogenetic activation of OVLT neurons significantly elevated renal, splanchnic, and lumbar sympathetic nerve activity. Third, single-unit recordings revealed optogenetic activation of the OVLT significantly increased the discharge of bulbospinal, sympathetic neurons in the rostral ventrolateral medulla. Lastly, chronic chemogenetic activation of OVLT neurons for 7 days significantly increased 24-hour fluid intake and mean arterial blood pressure. When the 24-hour fluid intake was clamped at baseline intakes, chemogenetic activation of OVLT neurons still produced a similar increase in arterial blood pressure. Neurogenic pressor activity assessed by the ganglionic blocker chlorisondamine was greater at 7 days of OVLT activation versus baseline. Collectively, these findings indicate that acute or chronic activation of OVLT neurons produces a sympathetically mediated hypertension.
Collapse
Affiliation(s)
- Sean D. Stocker
- Department of Neurobiology, University of Pittsburgh School of Medicine
| | - Megan M. Wenner
- Department of Kinesiology and Applied Physiology, University of Delaware
| | | | | |
Collapse
|
47
|
Firibastat: a Novel Treatment for Hypertension. Curr Hypertens Rep 2021; 23:46. [PMID: 34950965 DOI: 10.1007/s11906-021-01163-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to discuss the unique mechanism of firibastat, a new antihypertension medication. Hypertension continues to be a highly prevalent public health issue. RECENT FINDINGS Firibastat is a novel agent developed to treat hypertension. As the first member in the class of centrally acting agents to target the brain renin angiotensin system, firibastat offers new pathways to consider and enhances the regimen of agents currently available to treat hypertension. Recent clinical trials have demonstrated effectiveness and safety in mild hypertension as well as resistant hypertension. This review introduces firibastat as a new therapeutic class of treatment for hypertension focused on the renin angiotensin system in the brain. Early studies have shown a significant reduction in blood pressure with minimal side effects particularly in patients who are difficult to treat.
Collapse
|
48
|
Interplay between baroreflex sensitivity, obesity and related cardiometabolic risk factors (Review). Exp Ther Med 2021; 23:67. [PMID: 34934438 PMCID: PMC8649854 DOI: 10.3892/etm.2021.10990] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/03/2021] [Indexed: 11/26/2022] Open
Abstract
The baroreflex represents a rapid negative feedback system implicated in blood pressure regulation, which aims to prevent blood pressure variations by regulating peripheral vascular tone and cardiac output. The aim of the present review was to highlight the association between baroreflex sensitivity (BRS) and obesity, including factors associated with obesity, such as metabolic syndrome, hypertension, cardiovascular disease and diabetes. For the present review, a literature search was conducted using the PubMed database until August 21, 2021. The searched terms included ‘baroreflex’, and other terms such as ‘sensitivity’, ‘obesity’, ‘metabolic syndrome’, ‘hypertension’, ‘diabetes’, ‘gender’, ‘aging’, ‘children’, ‘adolescents’, ‘physical activity’, ‘bariatric surgery’, ‘autonomous nervous system’ and ‘cardiometabolic risk factors’. Obesity and its related metabolic disorders can influence baroreflex functionality and decrease BRS, mostly by potentiating sympathetic nervous system activity. Obesity induces inflammation, which can increase sympathetic system activity and lead to a higher incidence of cardiovascular events. Obesity also represents an important risk factor for hypertension through numerous mechanisms; in this setting, dysfunctional baroreceptors are not able to protect against constantly elevated blood pressure. Furthermore, diabetes mellitus and oxidative stress result in deterioration of BRS, whereas aging is also generally related to reduced cardiovagal BRS. Differences in BRS have also been observed between men and women, and overall cardiovagal BRS in healthy women is less intense compared with that in men. BRS appears lower in children with obesity compared with that in children of a healthy weight. Notably, physical exercise can increase BRS in both hypertensive and normotensive subjects, and BRS can also be significantly improved following bariatric surgery and weight loss. In conclusion, obesity and its related metabolic disorders may influence baroreflex functionality and decrease BRS, and baroreceptors cannot protect against the constantly elevated blood pressure in obesity. However, following bariatric surgery and weight loss, BRS can be significantly improved. The present review summarizes the role of obesity and related metabolic risk factors in BRS, providing details on possible mechanisms and shedding light on their interplay leading to autonomic neuropathy.
Collapse
|
49
|
Perrotta M, Carnevale D. Brain Areas Involved in Modulating the Immune Response Participating in Hypertension and Its Target Organ Damage. Antioxid Redox Signal 2021; 35:1515-1530. [PMID: 34269604 DOI: 10.1089/ars.2021.0142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Hypertension is a multifactorial disease ensuing from the continuous challenge imposed by several risk factors on the cardiovascular system. Classically known pathophysiological alterations associated with hypertension comprise neurogenic mechanisms dysregulating the autonomic nervous system (ANS), vascular dysfunction, and excessive activation of the renin angiotensin system. During the past few years, a considerable number of studies indicated that immune activation and inflammation also have an important role in the onset and maintenance of hypertension. Critical Issues: On these premises, it has been necessary to reconsider the pathophysiological mechanisms underlying hypertension development, taking into account the potential interactions established between classically known determinants of high blood pressure and the immune system. Recent Advances: Interestingly, central nervous system areas controlling cardiovascular functions are enriched with Angiotensin II receptors. Observations showing that these brain areas are crucial for mediating peripheral ANS and immune responses were suggestive of a critical role of neuroimmune interactions in hypertension. In fact, the ANS, characterized by an intricate network of afferent and efferent fibers, represents an intermediate between the brain and peripheral responses that are essential for blood pressure regulation. Future Directions: In this review, we will summarize studies showing how specific brain areas can modulate immune responses that are involved in hypertension. Antioxid. Redox Signal. 35, 1515-1530.
Collapse
Affiliation(s)
- Marialuisa Perrotta
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Daniela Carnevale
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy.,Research Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
50
|
Mowry FE, Peaden SC, Stern JE, Biancardi VC. TLR4 and AT1R mediate blood-brain barrier disruption, neuroinflammation, and autonomic dysfunction in spontaneously hypertensive rats. Pharmacol Res 2021; 174:105877. [PMID: 34610452 PMCID: PMC8648989 DOI: 10.1016/j.phrs.2021.105877] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/03/2021] [Accepted: 09/05/2021] [Indexed: 01/23/2023]
Abstract
Angiotensin II (AngII) is implicated in neuroinflammation, blood-brain barrier (BBB) disruption, and autonomic dysfunction in hypertension. We have previously shown that exogenous AngII stimulates Toll-like receptor 4 (TLR4) via AngII type 1 receptor (AT1R), inducing activation of hypothalamic microglia ex vivo, and that AngII-AT1R signaling is necessary for the loss of BBB integrity in spontaneously hypertensive rats (SHRs). Herein, we hypothesized that microglial TLR4 and AT1R signaling interactions represent a crucial mechanistic link between AngII-mediated neuroinflammation and BBB disruption, thereby contributing to sympathoexcitation in SHRs. Male SHRs were treated with TAK-242 (TLR4 inhibitor; 2 weeks), Losartan (AT1R inhibitor; 4 weeks), or vehicle, and age-matched to control Wistar Kyoto rats (WKYs). TLR4 and AT1R inhibitions normalized increased TLR4, interleukin-6, and tumor necrosis factor-α protein densities in SHR cardioregulatory nuclei (hypothalamic paraventricular nucleus [PVN], rostral ventrolateral medulla [RVLM], and nucleus tractus solitarius [NTS]), and abolished enhanced microglial activation. PVN, RVLM, and NTS BBB permeability analyses revealed complete restoration after TAK-242 treatment, whereas SHRs presented with elevated dye leakage. Mean arterial pressure was normalized in Losartan-treated SHRs, and attenuated with TLR4 inhibition. In conscious assessments, TLR4 blockade rescued SHR baroreflex sensitivity to vasoactive drugs, and reduced the SHR pressor response to ganglionic blockade to normal levels. These data suggest that TLR4 activation plays a substantial role in mediating a feed-forward pro-hypertensive cycle involving BBB disruption, neuroinflammation, and autonomic dysfunction, and that TLR4-specific therapeutic interventions may represent viable alternatives in the treatment of hypertension.
Collapse
Affiliation(s)
- Francesca E Mowry
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA; Center for Neurosciences Initiative, Auburn University, Auburn, AL, USA
| | - Sarah C Peaden
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Javier E Stern
- Center for Neuroinflammation, Georgia State University, Atlanta, GA, USA
| | - Vinicia C Biancardi
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA; Center for Neurosciences Initiative, Auburn University, Auburn, AL, USA.
| |
Collapse
|